Top Banner
1 Training Manual for Hemoglobinopathies and Hemophilia: Blood cell-MoHFW in collaboration with ICMR-National Institute of Immunohaematology Ministry of Health and Family Welfare, Government of India BLOOD CELL
442

Training Manual for Hemoglobinopathies and Hemophilia:

Mar 26, 2023

Download

Documents

Khang Minh
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Training Manual for Hemoglobinopathies and Hemophilia:

1

Training Manual for

Hemoglobinopathies and

Hemophilia:

Blood cell-MoHFW in collaboration with

ICMR-National Institute of Immunohaematology

Ministry of Health and Family Welfare, Government of India

BBLLOOOODD CCEELLLL

Page 2: Training Manual for Hemoglobinopathies and Hemophilia:

2

Page 3: Training Manual for Hemoglobinopathies and Hemophilia:

3

Page 4: Training Manual for Hemoglobinopathies and Hemophilia:

4

List of Contributors

Aby Abraham Professor, Dept. of Haematology, Christian Medical College, Vellore

[email protected]

Anita Harinkhede Senior Resident, Government Medical College, Nagpur

[email protected]

Anita Nadkarni Scientist F, ICMR-National Institute of Immunohaematology, Mumbai

[email protected]

Arihant Jain

Assistant Professor, Dept. of Internal Medicine, Post Graduate Institute of Medical Education andResearch, Chandigarh

[email protected]

Ashutosh Panigrahi

Assistant Professor, Dept. of Medical Oncology/Hematology, All India Institute of Medical Sciences, Bhubaneshwar

[email protected]

Bipin Kulkarni Scientist D, ICMR-National Institute of Immunohaematology, Mumbai

[email protected]

Chandrakala S Professor& Head, Department of Haematology, KEM Hospital, Mumbai

[email protected]

Dipti Jain Professor, Dept of Pediatrics, Government Medical College,Nagpur

[email protected]

Jasmina Ahluwalia

Professor, Dept. of Hematology, Post Graduate Institute of Medical Education andResearch,Chandigarh

[email protected]

Jayashri Kale Professor, Department of Occupational Therapy, KEM Hospital, Mumbai

[email protected]

Kanjaksha Ghosh Former Director, ICMR-National Institute of Immunohaematology, Mumbai

[email protected]

Maitreyee Bhattacharyya Professor, Dept of Haematology, Calcutta Medical College, Kolkata

[email protected]

Malay Mukherjee

Former Scientist F& Head, Dept of Hematogenetics, ICMR-National Institute of Immunohaematology, Mumbai

[email protected]

Manisha Madkaikar Director, ICMR-National Institute of Immunohaematology, Mumbai

[email protected]

Meenu Bajpai Addl. Professor, Transfusion MedicineInstitiute of Liver and Biliary Science,New Delhi

[email protected]

Page 5: Training Manual for Hemoglobinopathies and Hemophilia:

5

Nikesh Kawankar Technical assistant, ICMR-National Institute of Immunohaematology, Mumbai

[email protected]

Pallavi Mehta Technician C, ICMR-National Institute of Immunohaematology, Mumbai

[email protected]

Pankaj Malhotra

Professor, Dept. of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh

[email protected]

Prantar Chakravarty Former Professor, Dept. of Haematology, Nil Ratan Sircar Medical College, Kolkata

[email protected]

Prashant Sharma

Additional Professor, Dept of Hematology, Post Graduate Institute of Medical Education and Research,Chandigarh

[email protected]

Priya Hariharan Research Associate, ICMR-National Institute of Immunohaematology, Mumbai

[email protected]

Ravi Ranjan Scientist 1, AIIMS,New Delhi [email protected]

Reena Das

Professor, Dept. of Hematology, Post Graduate Institute of Medical Education andResearch,Chandigarh

[email protected]

Renu Saxena

Former Professor & Head Dept of Haematology, All India Institute of Medical Sciences, New Delhi

[email protected]

Richa Mohan

Clinical Psychologist, Empowering Minds Society for Research & Development, New Delhi

[email protected]

Roshan Colah Former Director In-Charge, ICMR-National Institute of Immunohaematology, Mumbai

[email protected]

Rucha Patil Scientist B, ICMR-National Institute of Immunohaematology, Mumbai

[email protected]

Sharda Shanbhag Technical assistant, ICMR-National Institute of Immunohaematology, Mumbai

[email protected]

Shrimati Shetty

Former Scientist F & Head Dept of Hemostasis, ICMR-National Institute of Immunohaematology, Mumbai

[email protected]

Shweta Sharma Dr. Shweta Sharma, Associate Professor, Dept of pediatrics, Gandhi Medical college, Bhopal

[email protected]

Soniya Nityanand

Professor& Head, Dept of Haematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow

[email protected]

Page 6: Training Manual for Hemoglobinopathies and Hemophilia:

6

Tulika Seth Professor, Dept of Haematology,All India Institute of Medical Sciences, New Delhi

[email protected]

V.P. Choudhry

Senior Consultant Hematology, Fortis Escorts Hospital, Batra Hospital & Medical Research Centre, Delhi,

[email protected]

Vikram Mathews Professor, Dept. of Haematology, Christian Medical College,Vellore

[email protected]

Vinita Srivastava

National Senior Consultant & Co-ordinator Incharge Blood cell - NHM, Nirman Bhavan, Ministry of Health & Family Welfare,GOI, New Delhi

[email protected], [email protected]

Page 7: Training Manual for Hemoglobinopathies and Hemophilia:

7

Contents

Section A: Hemoglobinopathies

Chapters Page

no.

Introduction 6

Government policy on hemoglobinopathies 11

Abbreviations 15

Definitions of Various Levels of Training 17

1. Laboratory Diagnosis of Hemoglobinopathies and Counseling

Screening andDiagnosis of Hemoglobinopathies 20

Newborn screening for Sickle Cell Disease 51

MolecularAnalysis of Hemoglobinopathies 62

PrenatalDiagnosis of Hemoglobinopathies 88

Quality Assurance and Equipment Maintenance in Hemoglobinopathies 100

Genetic Counseling for Hemoglobinopathies 124

2.Management of Hemoglobinopathies:

i.Thalassemia Major

Basic Recognition of Symptoms and Signs of Thalassemia Major at Level 1

(PHC/CHC)

133

Initial Work Up (See diagnostic section too), Stabilization and Triage of Care of the

Sick Patient

136

Guidelines for Blood Transfusion in Hemoglobinopathies 148

Guidelines for Chelation Therapy in Thalassemia 155

Monitoring and Data Collection 168

Common Complications and their Management in Thalassemia major 174

Transfusion Reactions 188

ii.Sickle Cell Disease

Basic Recognition of Symptoms and Signs of Sickle Cell Disease at Level 1

(PHC/CHC)

194

Important Guidelines for Management of Sickle Cell Disease 196

Hydroxyurea Therapy-Principles and Guidelines 202

Common Complications and their Management 209

Exchange Transfusion and Simple Transfusion in Sickle Cell Disease 227

Management of Newborn and Pregnanacy 235

Alloimmunization 242

Hematopoietic Stem Cell Transplantation (HSCT) for Thalassemia Major 247

Hematopoietic Stem Cell Transplantation (HSCT) for Sickle Cell Disease 254

Page 8: Training Manual for Hemoglobinopathies and Hemophilia:

8

Alpha Thalassemias: Pathophysiology and Diagnosis 261

Role of Genetic Modifiers 271

Newer Modalities for Management of Hemoglobinopathies 277

Registry for Hemoglobinopathies 284

Page 9: Training Manual for Hemoglobinopathies and Hemophilia:

9

Introduction

The inherited disorders of hemoglobin constitute a major global health problem and broadly

include the and thalassemias, symptomatic structural hemoglobinopathies like sickle

cell disease and Hb E as well as co-inheritance of these genes. They have attained high

frequencies in regions where malaria has been endemic. They are also the commonest group

of single gene disorders in India with an autosomal recessive inheritance.

The thalassemia syndromes and sickle cell disease and HbE pose the major health burden

in India. The average prevalence of thalassemia carriers is 3.5- 4% which translates to

around 40 to 50million people. In some communities the prevalence is higher varying from 5-

17 %. It has been estimated that 10,000 to 15,000 babies with a major thalassemia

syndrome are born each year in Inda. More accurate numbers will become available once

there is a registry of all patients and carriers. Sickle cell disease is prevalent in central India

and in parts of western, southern and eastern India mainly among tribal and some non-tribal

populations. The prevalence of sickle cell carriers in some of these groups can be as high as

25- 35%. Hb E is seen mainly in the north eastern region where the prevalence of Hb E

carriers is as high as 40- 50 % in some populations and Hb E is common in West Bengal.

Accurate diagnosis of the inherited hemoglobinopathies is important to confirm a suspected

clinical diagnosis for appropriate management of patients, explain hematological

abnormalities like anemia or microcytosis, to identify carriers or heterozygotes of

thalassemia or other abnormal hemoglobin variants who are generally asymptomatic but

require counseling and to identify couples at- risk of having a child with a severe disorder to

give them the option of prenatal diagnosis to avoid the birth of an affected child. Accurate

diagnosis of sickle cell disease is also crucial in the newborn period to reduce morbidity and

mortality by providing early care, intervention and regular follow up of these babies.

A combination of different methods as described in this manual should be used for postnatal

and prenataldiagnosis of these disorders and strict internal quality assurance (IQA) and

external quality assurance (EQA) must be undertaken for each of these hematological,

biochemical and molecular methodswhichare crucial to avoid any misdiagnosis.

Introducing screening programmes at the National level on a large scale is a big challenge

and would require adequate resources for prevention programmes and long-term care of

patients. Efforts should be intensified to strengthen awareness in the population.

Page 10: Training Manual for Hemoglobinopathies and Hemophilia:

10

Training of Health Professionals and other Health Workers at all levels must be undertaken

for all the states. Uniform protocols need to be followed throughoutthe country as described

in this training manual. All patients of the thalassemia syndromes and sickle cell disease

should have access to clinical mannagement and multidisciplinary care with recommended

monitoring and treatment protocolsas described in thistraining manual. Those patients who

are eligible and have a matched donor should have the possibility of cure by access to bone

marrow/ stem cell transplant.

Information along with guidance and support are required for a successful control

programme. In a large and diverse country like India, screening cannot be offered to

everyone, even if it is desirable and targeting selected groups initially would be profitable and

cost effective. Such groups include newly married couples and couples during early

pregnancy and extended family members of affected or carrier individuals. Premarital

screening is by and large not acceptable due to the social stigmatization attached to being a

carrier of thalassemia or another hemoglobinopathy.However, those who come voluntarily

should be screened. Prenatal diagnosis facilities would be established and offered to all

couplesin medical colleges in each state.

Past experience in many programmes has shown that once only advice is insufficient and

genetic counseling needs to be a continuous process for which adequate well-

trainedcounselors are required.

Managing and controlling these disorders nationally is largely the responsibility of both the

Central and State Governments. This Training Manual describes in detail the training to be

imparted at different levels in the Public Health System to reach out and benefit both the

urban and rural population. National Institutes in the country with expertise in diagnosis and

management of hemoglobinopathies will serve as Training Centres for medical colleges to

impart the highest level of training. The success of such a huge initiative at the public health

level depends critically on dealing effectively with managerial challenges, availability of

adequate infrastructure, resources and commitment of trained staff. Public-Private

partnerships can also be developed and different NGOs working in this area could be

involved particularly for awareness generation in the population. An Expert Advisory Group

would periodically review and monitor the progress of this Comprehensive National

Programme for Prevention and Management of Hemoglobinopathies in the country

Page 11: Training Manual for Hemoglobinopathies and Hemophilia:

11

Government Guidelines on Hemoglobinopathies

Thalassemia and Sickle Cell Disease are two common genetic disorders that are chronic, life-

restricting and require long and specialized treatment.They cause distress and financial loss to

the family and are a great drain on the health resources of the country. With the fall in Infant

Mortality Rate due to control of communicable and nutritional disorders in the last decade in

India, these disorders have become important causes of morbidity and mortality.It is

estimated thatin India there are almost 42 million carriers of β-thalassemia, around 1-

1.5million patients of Thalassemia major and about 10,000 to 15,000 babies with β-

thalassemia major are born each year. The carrier frequency of Sickle cell gene varies from 1-

35% and hence there are huge numbers of people with Sickle cell disease.

There are various challenges related to prevention and management of Hemoglobinopathies

in India. The epidemiological data is incomplete, and the precise burden of these disorders is

unknown due to lack of awareness and problems related to diagnosis in rural & remote

areas.Treatment of Thalassemia requires repeated blood transfusions which should be safe

blood to avoid the transmission of transfusion transmitted infections such as HIV, hepatitis B

and C.The excess iron that gets into the body through the blood transfusions needs to be

removed by use of the expensive chelator medicines. Bone marrow transplantation is a

curative treatment that requires aHLA-matched donor, specific infrastructure and trained

doctors and nurses. The physicians need specialized training to treat, monitor and manage

the complications of Thalassemia & Sickle Cell Disease.

Recognizing the socio-economic burden these disorders place on the family, society and the

health services, the Government of India has formed this policy guidelines aimed at

informing and providing broad guidance on prevention and management of these disorders.

A Technical Committee was constituted comprising of all stakeholders including experts,

patient groups; civil society organizations etc., to formulate the policy guidelines on

Haemoglobinopathies.This policy guidelines is based on the recommendations made by

Page 12: Training Manual for Hemoglobinopathies and Hemophilia:

12

thecommittee and from evidence provided by Indian & International medical literature along

with the Thalassemia International FederationGuidelines & WHO Guidelines.

This policy guidelines encompasses the public health goal of reducing the birth of affected

children through carrier screening and prenatal diagnosis and providing evidence- based

treatment for those affected.

An appropriate mechanism is recommended under Blood cell NHM at the National

and the State level for monitoring and implementation. Periodic review and mid-

course corrections based on new data and information would be carried out in

consultation with ICMR and DGHS.

Public Health and Hospitals is a state subject and therefore the policy guidelines is

meant to provide vision and broad guidance for prevention and management of

hemoglobinopathies tostates and UTs.

The guiding principle recognizes that for prevention, the focus should be on creating

awareness of these disorders in the community for acceptance of carrier screening,

which is recommended for all pregnant mothers, school & college going students.

Pregnant women identified to be carriers, and their husbands should be screened and

in couples, where both the partners are carriers, prenatal diagnosis should be offered

to ensure that the child is not affected with a clinically significant hemoglobinopathy.

Premarital and preconception carrier screening should be instituted with appropriate

genetic counseling. All subjects should be informed their status, whether normal,

carrier or diseased through systems of coding. For Sickle Cell Disease, the policy

guidelines recommends initiation of newborn screening in areas of high prevalence.

Those detected to have sickle cell anemia (HbSS) or compound heterozygosity of

HbS and β-thalassemia should be provided appropriate prophylaxis and

immunizations, especially pneumococcal and Hib vaccine, and followed up carefully

In the rural areas, ASHAs/multipurpose workers are envisaged to be trained and

utilized to identify individuals with severe anemia who couldhaveThalassemia major

or Sickle Cell Disease. These patients will be referred to the appropriate health care

facilities (District hospitals & above) for further testing and confirmation. The

district Hospitals/ Medical colleges should be equipped with equipment to measure

Page 13: Training Manual for Hemoglobinopathies and Hemophilia:

13

the hemoglobin, red cell indices, to carry out carrier screening of β-thalassemia and

sickle celldisease.

The strategy focuses on providing comprehensive services for patients with

Hemoglobinopathies by strengthening public health systems and establishing ICHH

centers in Districts/ Medical colleges. Italso envisages creation of Centres of

Excellence in state medical colleges with advanced facilities required for

comprehensive care of patients with Thalassemia/Sickle Cell Disease as well as with

the facility for prenatal diagnosis and hematopoietic stem cell transplant. Such

Centers of Excellence to function as hubs for providing technical support for those

situated at district and below level health facilities.

The program recommends creation of an integrated unit for Hemoglobinopathies and

Hemophilia keeping patient load in mind, in government medical colleges and district

level hospitals inaphased manner. The policy guidelines envisages a system of

referral from district hospital to medical college/ Center of Excellence (COE)

including use of digital technology such as tele- consultations. The document links the

Haemoglobinopathies programme with other programmeseg. Hepatitis, HIV and

Malaria programme.

The policy guidelines also recommends creation of a web-based application at the

state and National level for providing information in simple language with translation

in the common Indian languages, about the disease, its complications, their

management, and the places where different facilities are available.The guidelines

advocates a multi-stakeholder approach with participation of patients, parent support

organizations academic institutions, not for profit organizations, and health care

facilities.

The plan recommends setting up of a patient registry for Thalassemia and Sickle Cell

Disease to obtain information on the number of persons affected and the number of

carriers to estimate patients who require various services. The data on carrier

screening performed in different regions should be collated to determine the burden of

hemoglobinopathies.

Page 14: Training Manual for Hemoglobinopathies and Hemophilia:

14

The strategy advocates promoting research to develop innovative treatments for

Thalassemia major and Sickle Cell Disease, and devise new diagnostic methods,

keeping in mind the continuously evolving technology in this field.

Thus, the policy guidelines helps as a guiding tool to the concerned states and will

help in implementation of the programme in an improved manner

Abbreviations

AH Area Hospital

ANC Antenatal Clinic

ANM Auxiliary Nurse Midwife

Apo B Apolipoprotein B

ARMS Amplification Refractory Mutation System

ASHA Accredited Social Health Activist

AWW AnganWadi Workers

BMT Bone Marrow Transplantation

Bp Base Pair

BT Blood Transfusion

CBC Complete Blood Count

CCl4 Carbon tetrachloride

CHC Community Health Centre

DEIC District Early Intervention Centre

DH District Hospital

DMER Directorateof Medical Education and Research

DMSO Dimethyl Sulphoxide

DNA Deoxyribo Nucleic Acid

dNTP Deoxyribo Nucleotide Triphosphate

D/W Distilled water

Hb Hemoglobin

HbA0 Adult Hemoglobin

HbF FetalHemoglobin

HPLC High Pressure Liquid Chromatography

EQAS External Quality Assurance Scheme

HSCT Hematopoietic Stem Cell Transplantation

IEC Institutional Ethical Committee

IVS Intervening sequences

Kb Kilobase

LT Laboratory Technologist

MCH Mean CorposcularHemoglobin

Page 15: Training Manual for Hemoglobinopathies and Hemophilia:

15

MCHC Mean Cell HemoglobinConcentration

MCV Mean Corpuscular Volume

MO Medical Officer

NBS Newborn Screening

NESTROFT Nacked Eye Single Tube Osmotic Fragility Test

NTDT Non Transfusion Dependent Thalassemia

PCR Polymerase Chain Reaction

PHC Primary Health Centre

PND Prenatal Diagnosis

POC Point of Care

RBSK Rashtriya Bal SwasthyaKaryakram

RFLP Restriction Fragment Length Polymorphism

CRDB Covalent Reverse Dot Blot Hybridization

SCD Sickle Cell Disease

TAE Tris Acetate EDTA buffer

TBE Tris Borate EDTA buffer

TD Transfusion Dependent

TOT Training of Trainers

TDT Transfusion Dependent Thalassemia

VNTR Variable Number of Tandam Repeats

Page 16: Training Manual for Hemoglobinopathies and Hemophilia:

16

Definition of Various Levels of Training

Level 1:

Primary Health Centre (PHC)

Community Health Centre (CHC)

Level 2: Integrated Day Care Centre for Hemoglobinopathies and Hemophilia

District Hospitals and District Early Intervention Centres (DEIC)

Government Medical Colleges

Level 3:Centres of Excellence

Government Medical College (1 to 2): States to decide

Level 4: National Training Institutes

ICMR-National Institute of Immunohaematology (NIIH), Mumbai

All India Institute of Medical Sciences(AIIMS), New Delhi

Sanjay Gandhi Postgraduate Istitute of Medical Science (SGPGIMS),Lucknow

Christian Medical College (CMC), Vellore

Post Graduate Institute of Medical Education and Research(PGIMER), Chandigarh

Kolkata Medical College, Kolkata

States to be covered by each National Training Institute for training

Name of the Institute States to be covered

NIIH, Mumbai Maharashtra, Gujarat, Goa, Daman and Diu, Dadra and Nagar

Haveli, Karnataka

AIIMS, New Delhi Delhi, Rajasthan, Madhya Pradesh, Chattisgarh

SGPGIMS, Lucknow Uttar Pradesh, Bihar, Jharkhand

CMC, Vellore Tamil Nadu, Kerala, Andhra Pradesh, Telangana, Pondicherry,

Andaman and Nicobar

PGIMER, Chandigarh Punjab, Haryana, Himachal Pradesh, Uttarakhand, Jammu and

Kashmir

Medical College, Kolkata West Bengal, Orissa, Assam, Arunachal Pradesh, Meghalaya,

Sikkim, Manipur, Nagaland, Tripura

Level 1: Primary Health Centre (PHC)

Personnel: Training will be imparted to ASHA, ANM and RBSK worker at Level 2 by TOT

Training to be imparted: CBC on automated cell counter, preparation of peripheral smear,

staining and evaluation, solubility test, point of care testing for SCD, counseling.

Page 17: Training Manual for Hemoglobinopathies and Hemophilia:

17

CBC and peripheral blood smear examination should be done for all ANC women and

suspected cases of hemoglobinopathies and blood samples should be sent to level 2 at nearest

Integrated Centre for Hemoglobinopathies and Hemophilia.

Newborn Screening by Point of Care Testing for sickle cell disease. Any baby who tests

positive will be confirmed by HPLC and family screening will be done at level 3 (Centre of

Excellence).After confirmation of diagnosis, these babies will be registered in the Integrated

Centre for HemoglobinopathiesandHemophilia.

Instruments: Three-part automated blood cell counters and microscope

Level 2: IntegratedCentre for Hemoglobinopathies and Hemophilia to be established at

District Hospitals and Govt. Medical colleges.

Personnel: Training will be imparted to medical officers (MO), laboratory technicians (LT),

pathologists, counselors and obstetricians and training of trainers (TOT).

Designated MOs and staff at the District Hospital (DH) and District Early Intervention

Centres (DEIC) will be imparted training.

Training to be imparted: CBC on automated cell counter, preparation of peripheral smear,

staining and evaluation, solubility test, point of care testing for SCD, HPLC analysis

counseling.

Instruments:

Three-part automated blood cell counters and microscope

D-10 HPLC/Electrophoresis

Newborn Screening by Point of Care Testingfor sickle cell disease. Any baby who tests

positive will be confirmed by HPLC and family screening will be done at level 3 (Centre of

Excellence).After confirmation of diagnosis, these babies will be registered in the Integrated

Centre for HemoglobinopathiesandHemophilia.

Level 3: Centre of Excellence- Government Medical College (One or two per state)

Personnel: Social counselor, TOT trainer, Pathologist, Obstetrician for CVSand

cordocentesis, Clinical Hematologist, Molecular Biologist.

Approximately 2500 square feet with lecture theaters and laboratory space

forimpartingtrainingfor Level 1 and 2 participants will be required.

Training to be imparted: CBC on automated cell counter, preparation of peripheral smear,

staining and evaluation, solubility test, point of care testing for SCD, HPLC analysis

counseling. Molecular technologies for prenatal diagnosis for pathologists and molecular

biologists. Fetal tissue sampling (CVS), amniocentesis and cordocentesis for obstetricians.

Clinical evaluation and management protocols for the patients for clinical hematologists.

Page 18: Training Manual for Hemoglobinopathies and Hemophilia:

18

Instruments:

1. Five-part automated blood cell counters

2. HPLCVariant 2

3. HPLC NBS

4. Hb electrophoresis system for cellulose acetate membranes

5. Molecular Diagnosis: PCR machine, Submergedelectrophoresis system, Gel

documentation system, Automated DNA sequencer, Inverted microscope for CVS

dissection

Quality Control: Both External Quality Assessment (EQAS) and Internal Quality Control

(IQC) to be done regularly.

Concept of Reagent Rental with maintenance of equipmentcould be used which will include

EQAS.

Level 4: National Training Institutes: NIIH, Mumbai; AIIMS, New Delhi; SGPGI,

Lucknow; CMC, Vellore; PGIMER, Chandigarh; Medical College Kolkata, Kolkata

Instruments:

1. Five-part automated blood cell counters

2. HPLC: Variant 2

3. Isoelectric focussing

Molecular Diagnosis: PCR machine, Submergedelectrophoresis system, Gel documentation

system, Automated DNA sequencer, Inverted microscope for CVS dissection.

Linkage Plan of action between all the Levels (1-4) will be assured by the National Training

Institutes.Level 4 will train the staff of level 3. Later Level 3 will train the staff of Levels 1

and 2. Patients from level 2 may be given preferential care and management at levels 3 and 4.

Voluntary thalassemia screening will be offered in all the centres

Additional staff to be provided as mentioned in the Guidelines at different levels by the

State/GOI

Page 19: Training Manual for Hemoglobinopathies and Hemophilia:

19

1. Laboratory Diagnosis of Hemoglobinopathies and Counseling

Screening and Diagnosis of Hemoglobinopathies

Introduction The hemoglobinopathies comprise of the inherited disorders of the structure or synthesis of

hemoglobin. They are one of the commonest groups of single gene disorders in the Indian

subcontinent and pose a major concern on our health resources.

The hemoglobinopathies can be broadly classified into three groups.

1. Hemoglobin variants, where there is a structural alteration in one of the globin chains

e.g.HbS, HbE, HbC, HbD etc.

2. The thalassemias, where there is impaired synthesis of normal globin chains

e.g.thalassemia, thalassemia, thalassemia, thalassemia and thalassemia.

3. A diverse group of conditions, in which there is a defect in developmental progression

from fetal to adult hemoglobin production e.g. hereditary persistence of fetalhemoglobin

(HPFH).

The abnormal hemoglobinsresultfrom mutations in the α, , or δ globin genes resulting

mainly in single amino acid substitutions. Both the thalassemias and abnormal hemoglobins

can be co-inherited to give compound heterozygous conditions.

Human hemoglobin is heterogenous at all stages of development, beginning with the

youngest embryos that have been studied and continuing throughout adult life. These

hemoglobins are designated by the Greek letters alpha (α), beta (β), gamma (γ), delta (δ),

epsilon (ε) and zeta (ζ). In embryos, hemoglobin synthesis is confined to the yolk sac, where

hemoglobin Grower 1 (22), Grower 2 (α22) and Portland (22) are produced. Synthesis of

chains becomes detectable at about 6th

weeks of gestation, when it comprisesof nearly 1.5

% of the non alpha chains, increasing to 5% at the 7th

week and nearly 10% by the 10th

week.

At around 7-8 weeks of gestation the liver becomes the major site of erythropoiesis,

producing large enucleated red cells. Throughout most of fetal life HbF production

predominates, with a small amount (<10%) of adult hemoglobin (HbA). The different

chains are produced in a ratio of G to

A of 3:1, which remains constant until late in

gestation. At midterm the bone marrow begins to take over as the major site of red cell

Page 20: Training Manual for Hemoglobinopathies and Hemophilia:

20

production, though erythropoiesis is also found in the spleen, as well as in other tissues.

Towards the end of gestation there is a gradual and reciprocal switch from HbF to HbA

production. At birth, cord blood normally contains nearly 70% HbFwhich declines to nearly

20% by 3 months, 7.5% at 6 months, and less than 2% by the age of oneyear. At the same

time there is a differential decline in G and

A chain production. Both fetal and adult

hemoglobins are produced in the same cell during the switching period, with gradual increase

in the production of cells containing predominantly HbA (Figure1).

Figure1: Developmental changes in hemoglobin production. The sites of erythropoiesis

during development and different globins produced at each stage.

(http://en.wikipedia.org/wiki/Hemoglobin)

Table 1: Different hemoglobins present in normal newborn and adults are given.

HbA0 HbA2 HbF

Newborn 20-30% 0-1.5% 50-80%

Adult 95-98% 2-3.0% 0.8-2%

As per the Hbvar database (http://globin.cse.psu.edu/hbvar/menu/html) there are more than

450 mutations causing the thalassemias and more than 1000 hemoglobin variants. Many

Page 21: Training Manual for Hemoglobinopathies and Hemophilia:

21

variants of hemoglobin are harmless whereas some have significant clinical presentation.

Many of the Hb variants are picked up during population screening programmes; however,

some are identified while investigating cases of microcytosis, hemolyticanemia, cyanosis or

erythrocytosis. Few variants are unstable and may result in a thalassemia intermedia

phenotype.

Thalassemia minor individuals (carriers of thalassemia) are usually clinically asymptomatic

but occasionally they may have mild to moderate anemia. Children born with thalassemia

major usually develop the symptoms of severe anemia within the first year of life and are

unable to produce normal adult hemoglobin. They are chronically fatigued, fail to thrive, do

not grow normally and prolong anemia causes bone deformities.

All hemoglobinopathes are inherited in an autosomal recessive manner. If both parents are

thalassemia carriers, their children may be thalassemia carriers, or completely normal, or they

may have thalassemia major. In each pregnancy there is a one in four (25%) chance that their

child will be normal, a two in four (50%) chance that the child will have thalassemia minor or

a one in four (25%) chance that the child will have thalassemia major. Diagnosis and

management of these disorders both in the neonatal period or later using appropriate

approaches and uniform technology are extremely important.

The basic laboratory diagnoses for identification of thalassemia carriers are red cell indices

and morphology followed by hemoglobin electrophoresis. The cut-off value of HbA2 for

diagnosis of β-thalassemia carriers is generally taken as 3.5% along with the presence of

reduced red cell indices (MCV < 80 fl and MCH< 27 pg) and a relatively high RBC count

and normal RDW values. In α-thalassemia, HbA2can be lower than normal and it is

significant when iron deficiency is excluded.

The β-thalassemias and the common β chain variants are easily identified using an automated

HPLC machine. The atypical and or unusual ones require further investigations like

hemoglobin electrophoresis at alkaline and acidic pH, tests for hemoglobin stability like heat

stability and isopropanol stability test, oxygen dissociation curve and spectral analysis. Often

DNA sequencing of the respective globin genes is required for identification of the variant.

The common β-chain variants in India are Hb S, HbE and HbDPunjab

. Hemoglobin S is

predominantly seen in tribal and in a few nontribal populations in central, western and some

parts of eastern and southern India with the prevalence of heterozygous going as high as 35-

Page 22: Training Manual for Hemoglobinopathies and Hemophilia:

22

40% in some groups. Hemoglobin E is very common in the north-eastern region where

carrier frequencies go up to more than 50% in some groups. HemoglobinDPunjab

is mainly

seen in north-western India where the prevalence varies from 1-3%. Many clinically

significant α-chain variants are also seen amongst the Indian population which includes

HbQIndia

, HbSallanches, Hb Sun Prairie, Hb Evanston, Hb Jackson, Hb O Indonesia, Hb J

Paris (I), Hb J Meerut,

HbKoyaDora, Hb Hofu, and Hb Fontainbleau. Among these HbQIndia

is commonest and seen

mainly in the Sindhi community.

Individuals to be screened

All pregnant women (preferably in thefirst trimester but also those who come

later).

Husbands of all pregnant women who are carriers (heterozygous) of β-

thalassemia, Hb S, Hb E, δβ thalassemia.

Husbands of all pregnant women who are homozygous/compound

heterozygous for the following: HbShomozygous, HbS-β-thalassemia, HbE

homozygous, HbE-β-thalassemia, HbD-β-thalassemia and HbQ-β-thalassemia.

Extended family members of β-thalassemia major, Sickle cell disease patients

and siblings of carriers identified during screening.

All individuals who request for screening voluntarily.

High risks communities should be screened.

All couples where both husbands and wives are carriers should be referred for

molecular analysis and counseling with the option of prenatal diagnosis to be done at

the nearest Center of Excellence.

Page 23: Training Manual for Hemoglobinopathies and Hemophilia:

23

Investigations to be performed for diagnosis of the patients

CBC with peripheral blood smear examination

Reticulocyte count

HbH inclusion bodies and Heat/ Iso-propanol stability if required (If

suspecting HbH disease and unstable hemoglobin)

Solubility test

HPLC

Refer to center of excellence for molecular confirmation

For confirmation of alpha thalassemia and rare hemoglobin variants refer to

National Institutes.

Recommendations on Technologies to be used for screening

CBC to be done for all pregnant women and all the other groups of

individuals screened.

Along with CBC, solubility test for sickle hemoglobin to be done in regions

where HbS is prevalent.

All individuals with MCV< 80fl, MCH< 27pg and/or a positive test for HbS

require HPLC analysis which is recommended for diagnosis.

These positive samples should be sent to the Integrated Centre for

Hemoglobinopathiesand Hemophilia for HPLC for diagnosis

Hb electrophoresis on cellulose acetate at alkaline pH should be done to

differentiate hemoglobins which elute in the same window in HPLC (Hb

Lepore, HbE and HbDIran

)

Classical β-thalassemia carriers will have MCV<80, MCH<27, RDW -Normal, RBC

Count - Increased, HbA2> 3.5% and HbF 0.5-2.0%

Page 24: Training Manual for Hemoglobinopathies and Hemophilia:

24

Complete Blood Count

Principle: Complete blood count to determine red blood cell (RBC) indices is the most

common laboratory test and is usually carried out using blood collected in EDTAon an

automated electronic cell counterwithin a few hours of blood collection. Parameters such as

hemoglobin (Hb) concentration, mean corpuscular volume (MCV), mean corpuscular

hemoglobin (MCH), RBC count and red cell distribution width (RDW) are strictly relevant

and useful for hemoglobinopathies screening (Table 2). Several automated cell counters are

available for doing a complete blood count. These counters work on the principle of electrical

impedance or laser light scattering. The counter needs to be calibrated daily with appropriate

material to obtain accurate results.

Interpretation:

MCV and MCH are variably reduced in thalassemia carriers. MCH is more reliable

than MCV, since the MCV does not remain stable due to a tendency for the red cells

to increase in size over timeduring storage.

The most widely used cut-off values of MCV and MCH for suspecting the presence

ofthalassemia carriersareMCV <80 fl and MCH <27 pgrespectively.

Silent β-thalassemia carriers may have normal MCV and MCH values.

RBC count is usually on the higher side in relation to the hemoglobin level in both

and thalassemia carriers.

RDW is normal in thalassemia carriers.

α-thalassemia carriers also have reduced MCV and MCH. However, α-thalassemia

carriers having asinglegene deletion may have near normal indices.

δβ-thalassemiacarriers have slightly reduced MCV and MCHvalues.

Page 25: Training Manual for Hemoglobinopathies and Hemophilia:

25

Table 2: Hematological parameters in thalassemia carriers and iron deficiency anemia.

Parameters with normal

range thalassemia

carriers

thalassemia

carriers

Iron deficiency

anemia

MCV (80-95 fl) Reduced Reduced Reduced

MCH (27-34 pg) Reduced Reduced Reduced

RBC count (4.5-5.6x 1012

/L) Increased Increased Reduced

RDW (11.5-14.5%) Normal Normal Increased

Red Blood Cell Morphology (Peripheral blood smear)

Principle: Morphological changes of red cells can be detected in most thalassemia carriers.

Theexamination of astained peripheral blood smear may be helpful in the evaluation of the

cases. Romanowsky stains are used for staining blood films. These are formulated by

blending methylene blue and eosin. Methylene blue is converted to it’s active form methyl

azures; when mixed with eosin, these stains are designated as polychromes because they

impart metachromatic qualities to the cell constituents.

Preparation of smear: EDTA anticoagulatedvenous blood or capillary blood is used to

prepare a blood film.

Reagents:

Leishman’s Stain:

0.2 g powdered Leishman’s dye is added to 100 mL methanol (Acetone free) and the

mixture is warmed to 500C in a shaking water bath for 15 minutes.

The solutionis then filtered and allowed to stand at room temperature for 24 hours.

This is stored at room temperature (250

C) in a dark bottle.

A pH of 6.8 is recommended for general use.

Method:

1. A small drop of blood is smearedon theslide using a spreader slide at an angle of 450.

Page 26: Training Manual for Hemoglobinopathies and Hemophilia:

26

2. The blood smear is driedat room temperature. Adequate drying is essential to preserve

the quality of the film.

3. By using alead pencil, the identification number is writtenon the slide.

4. The smear is covered with the staining solutionfor 2 minutes.

5. Distilled water is then added on the slide and the reaction mixture adequately

mixedby blowing on it using a pipette and allowed to standfor 20 minutes.

6. The slide is then rinsed in running tap water.

7. PBFis allowed to air dry and observed under theoil immersion lens of the microscope

(Figure 2).

Page 27: Training Manual for Hemoglobinopathies and Hemophilia:

27

Interpretation:

Figure 2: Red cell morphologyof a thalassemia carrier showing

anisocytosis,poikilocytosis, hypochromia and microcytosis

Microcytosis, hypochromia and anisopoikilocytosis are most typical changes in

thalassemia.

Other less common findings are basophilic stippling and presence of some target

cells.

Nucleated RBCs are indicative of bone marrow hyperactivity and can be found in

homozygous β-thalassemia.

Polychromasia is associated with the presence ofreticulocytosis.

Howell-Jolly bodies can be found after splenectomy or in the functional asplenic

condition in sickle cell syndromes, where sickle shaped cells are sometimes seen on

the stained films as well.

Reticulocyte Count and Hb H Inclusion Body Detection

Principle: For the detection of α-thalassemia, especially HbH disease, new methylene blue

stain will detect the characteristic HbH inclusion bodies. New methylene blue being a

supravital stain is able to stain residual mRNA in immature red blood cells (Reticulocytes).

Sample: EDTA anticoagulated blood

Page 28: Training Manual for Hemoglobinopathies and Hemophilia:

28

Reagents:

Iso-osmotic Phosphate buffer:

A. NaH2PO4.2H2O (150mM/L): 2.34g is dissolved in 100 mL of distilled

water (D/W)

B. Na2H PO4 (150mM/L): 2.13g is dissolved in 100 mL D/W

Note: Add Solution A- 18.0 mL and Solution B- 82 mL to prepare 100

mL of Iso-osmotic Phosphate buffer.

New Methylene Blue (NMB) stain: 1.0 gm (NMB) is dissolved in 100 mL of

Iso-osmotic phosphate buffer (pH- 7.4). The mixture is filtered and used.

Method:

1. Two drops of blood aremixed with one drop of stain in a tube and incubated at 370C

for 20 to 30 minutes.

2. A smear is then made on a grease free slide, air dried and observed under the oil

immersion lens of a microscope.

3. Reticulocytes are identified by their deep blue reticulated appearance.

4. A minimum of 1000 RBCs in successive fields are counted to determine the

percentage of reticulocytes.

5. Inclusion bodies can also be seen if the mixture of blood and dye is incubated for 1

hour at 370C and can be seen under the microscope (Figure 3)

Calculation:

Normal Values:

Adults and children: 0.2- 2.0 %

Newborn babies (cord blood/ full term baby): 2.0- 6.0 %

Page 29: Training Manual for Hemoglobinopathies and Hemophilia:

29

Observation:

Figure 3: Staining for A. reticulocytes and B. HbH inclusion bodies.

Sickling Test (For detection of HbS)

Principle: Sickling test is a part of the diagnostic workup in patients suspected of having a

sickle cell syndrome. Sodium metabisulphite reduces the oxygen tension inducing the typical

sickle shape of red blood cells.

Sample: Freshly collected EDTA anticoagulated blood

Reagents:

2% Sodium metabisulphite solution:0.2 g sodium metabisulphite is dissolved in 10.0

mL of distilled water. This solution should be prepared freshly.

Method:

One drop of blood is mixed with 3 drops of 2% sodium metabisulphite solution on a

glass slide.

A cover slip is placed over it and sealed with petroleum jelly or nailpaint.

The slide is placed in a petri dish, kept moist with wet cotton plugs and incubated at

370C for 1 to 4 hours.

The slide is observed under thehigh power(40X)lens of the microscope for the

presence of sickled cells (Figure 4).

Occasionally the preparation may need to stand up to 24 hours.

A B

Page 30: Training Manual for Hemoglobinopathies and Hemophilia:

30

Interpretation:

Figure 4: Sickling test showing typical sickle shaped cells.

Solubility Test

Principle:It is a rapid method for detection of HbS. HbS is quite insoluble when in the

reduced state in ahigh phosphate buffer solution. It forms water crystals (Tactoids) which

refract and deflect light rays and producea turbid solution.

Reagents:

Phosphate Buffer (pH 7.1): Stock Solution

Potassium dihydrogen phosphate (KH2PO4): 125.0 g

Dipotassium hydrogen phosphate (K2HPO4): 217.0 g

Saponin : 2.5 g

Distilled water : 1 L

Preparation of working solution: 0.1 g of sodium dithioniteis added to 10 mL of the

stocksolution immediately before use.

Method:

1. The RBCs are washed three times with normal saline.

2. Two mL of the phosphate buffer is takenand 20 µL of packed RBCsare added.

3. The tube is mixed well to give a light violet color.

4. The results are recorded after 10 minutesby holding a white paper with a dark black

line behind the tube (Figure 5).

Page 31: Training Manual for Hemoglobinopathies and Hemophilia:

31

Interpretation:

Positive result (+): the black line is not visible.

Negative result (-): the black line is clearly visible.

Doubtful result (+/-): the black line is partially visible.

Figure 5: Solubility test for Hb S detection

Preparation of Hemolysate for Hb Electrophoresis and Screening for

UnstableHemoglobins

Hemolysate preparation is required for two tests (Hb stability test and cellulose acetate

electrophoresis test) for detection of abnormal hemoglobin variants. For both the tests, the

initial step of red cell washing with saline is common. The difference lies in the addition of

distilled water to the packed RBC pellet. This is followed by the addition of Carbon

tetrachloride (CCl4) which is same for both the tests.

Requirements

Plastic tubes

Normal saline

Distilled water

CCl4

Centrifuge

Method

1. 1000 µL of blood is taken in a 1.5 mL tube and washed thrice with 0.9 % saline.

Page 32: Training Manual for Hemoglobinopathies and Hemophilia:

32

2. Lysate for heat stability test: 200 µL of packed RBCs are mixed with 400 µL of

distilled water in a fresh tube.

3. Lysate for Cellulose acetate electrophoresis test: 200 µl of packed RBCs are mixed

with 30 µL (2 drops) of distilled water in a fresh tube.

4. Tubes are vortexed thoroughly for 20 seconds to lyse the cells.

5. 200 µL of CCl4 is added to the above mixture to remove the cell membranes and other

cell debris.

6. The tubes are vortexed on a vortex mixer for 2 minutes and centrifuged at 10000 rpm

for 15 minutes.

7. The clear hemolysate from the top is pipetted out leaving behind the CCl4 (bottom

layer) and the cell membrane layer (middle layer).

Comments

Toluene can also be used instead of CCl4 in which case the hemolysate will be in the bottom

layer and pipetting out the lysate may be more difficult.

The hemolysate can be preserved at -200C after addition of 1-2 drops of 1 % potassium

cyanide (KCN) for cellulose acetate electrophoresis. Freshly prepared lysate without KCN is

used for the Hb stability test.

HemoglobinStability Test

Disruption of normal structure of the hemoglobin molecule can result in reduced stability,

which leads to precipitates in the erythrocyte causing it’s destruction. Amino acid

substitutions in the globin chains, particularly those involving non-polar amino acids that

constitute the heme pocket, may result in an unstable hemoglobin. However, if the substituted

amino acid is internal or the total charge of the molecule is unchanged, the hemoglobin

variant will not be detected by conventional electrophoresis. Therefore, if

anunstablehemoglobin is suspected clinically, a specific test for detection of the unstable

hemoglobin should be performed. Milder instability may also beassociated with Hb H

andHbvariants with altered oxygen affinity.

There are two stability testsIsopropanol stability test and Heat stability test.

Page 33: Training Manual for Hemoglobinopathies and Hemophilia:

33

Isopropanol stability test

Principle: The presence of isopropanol makes the buffer less polar, weakening the

hemoglobin hydrophobic binding that facilitate it’s denaturation and precipitation.

Reagents:

0.1 M Tris –HCL

12.1gmTris is dissolved in 600 mL distilled water, the pH is adjusted to 7.4 with 10 N

HCl and the volume made up to 1 liter.

17% Isopropanol- Tris buffer

17 mL of isopropanol is mixed with 83 mL of Tris-HCl buffer (pH 7.4). this buffer is

stable at room temperature.

Method:

1. 2 mL of isopropanol buffer is taken in a tube and equilibrated at 370C in a water bath

for 5 minutes.

2. 200 L of a freshly prepared hemolysate is then added and mixed properly. The tube

isagain re-incubated at 370C.

3. The tube is observed every 5 minutes to look for precipitation. For the unstable

hemoglobin the precipitate will form after 20 mins (Figure 6)

4. A normal control lysate is always put up simultaneously.

Interpretation:

Figure 6: Isopropanol stability test for unstable hemoglobin detection.

1. Normal Control: Clear solution after 1 hour of

incubation at 50oC.

2. Test sample: The solution turns turbid and a

flocculent precipitate forms within 20 minutes at

37oCwhich precipitates.

Precipitation

Page 34: Training Manual for Hemoglobinopathies and Hemophilia:

34

Heat stability test

Principle: Normal hemoglobin precipitates only slightly when incubated at 500C for 30

minutes, while an unstable hemoglobin under these conditions is completely denatured.

Sample: EDTA anticoagulated blood.

Reagents:

0.05 M Tris-HCL buffer

6.05gmTris is dissolved in about 600 mL distilled water. The pH is adjusted to 7.4

with 10 N HCl and the volume is made up to 1 liter.

Method:

1. 1.8 mL of 0.05 M Tris- HCL buffer is taken in a Kahn tube and equilibrated at 500C

in a water bath for 5 minutes.

2. 200 L of a freshly prepared hemolysate is then added and mixed properly. The tube

is re-incubated at 500C.

3. The tube is observed every 5 minutes to look for precipitation till 60 minutes.

4. A normal control lysate is always put up simultaneously.

Interpretation:Same as for isopropanol stability test

Hemoglobinpattern analysis

Different patterns of hemoglobin can be detected by using an electrophoretic method

(Cellulose acetate electrophoresis) and/or automated chromatographic methods (analysis on

D10 andHPLC Variant 2).

Cellulose Acetate Electrophoresis

Principle: Electrophoresis is a separation technique based on the mobility of ions in an

electric field. It is a classical method of identifying and quantifying the hemoglobin proteins.

At alkaline pH (8.4 to 8.6), hemoglobin is a negatively charged protein and migrates towards

Screening for unstable hemoglobin to be done at level 3 if required

Page 35: Training Manual for Hemoglobinopathies and Hemophilia:

35

the anode in an electrical field. During electrophoresis, various hemoglobins separate due to

charge differences caused by structural variations, thereby allowing their identification.

Cellulose acetate membranes demonstrate several features making them superior to filter

paper for hemoglobin separation. The absence of adsorption yields clean white backgrounds

between fractions and there is no adsorptive loss of protein during migration, hence theycan

be used to quantitate hemoglobinfractionson a spectrophotometer. The electro-endosmotic

flow is greater than in filter paper. This results in a continuous flow of buffer towards the

cathode and better separation of bands.

Reagents and materials:

Cellulose acetate membranes (stored in 30% methanol)

Tris EDTA Boric acid(TEB) buffer, pH 8.6

Tris : 14.4 g

EDTA (di sodium) : 1.5 g

Boric acid : 0.9 g

Distilled water to makethevolume to 1 L.

0.2% Ponceau S stain: 0.2 g of Ponceau S stain is dissolved in 100 mL of 3%

trichloroacetic acid

Destaining solution (2% Glacial acetic acid): 2 mL acetic acid made to 100 mL with

distilled water.

Paint brush

Horizontal electrophoresis tank

Filter paper wicks

Method:

1. Hemolysate is prepared from anticoagulated blood using theearlier mentioned

method.

2. The cellulose acetate membrane strips are soaked in TEB buffer for 30 minutes.

3. Excess buffer is removed by blotting the membranes between Whatman no. 1 filter

paper.

4. The membranes are placed across the bridge ofahorizontal electrophoresis chamber.

5. The membranes are secured using a double layer of Whatman no. 1 filter paper as

wicks. These wicks dip in the anode and cathode buffer compartments of the

chamber.

6. Hemolysate is applied at the cathode end of the strip using a fine tipped paint brush.

Page 36: Training Manual for Hemoglobinopathies and Hemophilia:

36

7. Electrophoresis is carried out at a constant voltage of 200-250 V (~ 1 mA/strip) for

1½ to 2 hours till adequate separation of bands is obtained.

8. The membranes are stained for 1 minute with Ponceau S stain in a petri dish and

destained in 2 % acetic acid till the background is clear.

9. The membrane can then be preserved in the destaining solution.

10. During every run it is advisable to electrophorese a known sample having a variant

hemoglobin as a control (Figure 7).

Interpretation:

Figure 7: Cellulose acetate electrophoresis at alkaline pH (8.9) (CA is carbonic

anhydrase)

The hemoglobins migrate on the cellulose acetate membrane from cathode to anode in the

following order: HbA2/ HbE, HbC, HbD/HbS, HbLepore, HbF, HbA0 and the fast

movinghemoglobin Bart’s and HbH.

HPLC for screening for hemoglobinopathies

Automated Cation exchange high performance liquid chromatography (CE-HPLC)

Principle: This method has emerged as the method of choice for quantification of HbA2,

HbF and for detection and quantitation of the Hb variants, particularly those which may

interact with β-thalassemia such as HbS, HbE, HbDPunjab

and Hb-Lepore. The Variant II

machineor the D10machine from BioRad laboratories has most commonly been used in

India, however, any other similar HPLC machine can be used. In this method phosphate

buffersof different concentration (mobile phase), pass under pressure through an ionic

+ -

N

HbA + HbS/D Punjab

HbS + HbF

HbE + HbF

Page 37: Training Manual for Hemoglobinopathies and Hemophilia:

37

exchange column (stationary phase). The stationary phase consists of a temperature

controlled analytical cartridge containing a resin of anionic or cationic particles (3-5 μm).

Two pumps and apre- programmed gradient control the elution buffer mixture (mobile phase)

passing through the analytical cartridge (stationary phase). As the ionic strength of the elution

buffer mixture increases, more strongly retained hemoglobins elute from the cartridge. A

photometer monitors the eluate and detects absorbance changes at 450 nm. Background

variations are compared by usingan additional filter at 690 nm. A chromatogram of

absorbance v/s time is displayed and printed. Windows are set for common hemoglobin

variants based on their retention times. Each sample takes 6.5 minutes for analysis and up to

100 samples can be loaded in the sampling chamber.

Sample:EDTA anticoagulated blood.

Analysis on D10 HPLC machine

Reagents:

Preparation of specimen

EDTA blood sample

If the blood sample is <2mL: 1.5 mL of wash/diluent solution and 5 µL of the

blood are mixed and used.

If the blood sample is≥ 2 mL: direct vacutainers can be placed in the machine.

Preparation of Primers

Reconstitute lyophilized primer with 1 mL of distilled water.

Allow to stand for 10-15 minutes; swirl gently to dissolve.

Stable for 1 day at 2-80C.

Preparation of the calibrator

Two calibrators (Level 1 and Level 2)

Reconstitute each vial with 7 mL of cold Calibrator Diluent.

Allow to stand for 5-10 minutes; swirl gently to dissolve.

Stable for 10 days at 2-80C.

Preparation of Controls

There are two sets of controls available (Level 1 and level 2)

Reconstitute each vial with 0.5 mL of distilled water

Allow to stand for 5-10 minutes; swirl gently to dissolve

Controls are stable for 21 days at 2-80C

Page 38: Training Manual for Hemoglobinopathies and Hemophilia:

38

Dilute 5µL of control in 1.5 mL of wash/Diluent solution.

Method:

1. Switch on the machine.

2. Place the system in sleep state

3. Go to LOT INFOscreen

4. Press the method box to display the select method screen.

5. Select A2/F METHOD

6. Press EXIST

7. Press YES to confirm the method change.

8. Press EXIT

9. The selected method is indicated in the status bar.

10. There is no need to perform a system flush unless a different lot of reagent is

installed.

11. A priming run is performed once per new cartridge and also following the

decontamination procedure.

12. Follow the instruction manual which comes along with the kit for priming.

13. Calibration is performed once when a new cartridgeis used.

14. Follow the instruction manual which comes along with the kit for calibration.

15. The calibration report is printed after the testing is complete. The slope and intercept

acceptable ranges are provided in calibrator diluents set insert.

16. Once the calibration is passed you can run the sample.

17. Once the cartridge has been calibrated there is no need to calibrate the cartridge again

when you switch on the machine next time.

18. Two different controls Level 1 and Level 2 are to be run every time before patient’s

samples.

19. Each sample will take 6.5 mins. to run.

Interpretation: (Figure 8)

HbA2<3.5%: Normal

HbA2 3.6-3.9 %: Borderline HbA2[needs confirmation by DNA analysis]

HbA2> 4.0 %: β-thalassemia trait (BTT)

HbA2< 2.0 %: Can be α-thalassemia / δ-thalassemia heterozygote

Common Hb variants such as HbSare identified as a separate peak.

Page 39: Training Manual for Hemoglobinopathies and Hemophilia:

39

HbDPunjab

andHbQIndia

elutes as an unknown peak after HbA2witharetention time of 3.83 min

and4.37 min respectively

Page 40: Training Manual for Hemoglobinopathies and Hemophilia:

40

Figure 8: HPLC chromatograms on D10 showing different hemoglobinopathies.

Page 41: Training Manual for Hemoglobinopathies and Hemophilia:

41

Analysis on Variant II HPLC machine

Reagents:

Preparation of specimen

EDTA / heparinised blood sample

If the blood sample is < 2 mL: 1mL of hemolysing solution and 5 µL of the

blood are mixed and used.

If the blood sample is≥ 2 mL: direct vacutainers can be placed in the machine.

Preparation of the Primer

1 mL of distilled water is addedto the lyophilized primer and mixed properly.

It is allowedto stand at room temperature for 10 minutes at 15-300C.

Preparation of the calibrator

10 mL of calibrator diluent is added to the lyophilized calibrator whichcomes

with the kit. It ismixed properly and allowed to stand at room temperature for

10 minutes at 15-300C.

The reconstituted calibrator is stable for 10 days when stored in aliquots at 2-

80C.

Aliquotsare made of this prepared calibrator.

Preparation of the controls

A set of normal (HbF: 1-2%, HbA2: 1.8-3.2%) and abnormal (HbF: 5-10%,

HbA2: 4-6%) controls are to be prepared according to the instructions.

The controls should be run at the beginning of each group of test specimens.

Limitations:

HbLepore, HbE and HbD Iran are co-eluted with HbA2.

HbA2is falsely increased in thepresence of HbS.

HbA2 is falsely reduced in thepresence of HbD.

HbDPunjab

elutes in anunknown window

Elevated HbF in case of β-thalassemia major and intermedia will elute in two

windows A1b and LA1C/CHb. In such acase,you need to add the concentration of

Hb from both the windows to get thetotal HbFlevel.

Hb Bart’s show a sharp peak at the start of the chromatogram and HbH shows twin

peaks before 1 minute.

Page 42: Training Manual for Hemoglobinopathies and Hemophilia:

42

Method:

1. The machine is first switched onfollowed by the computer.

2. The software is startedby clicking on the CDM icon

3. The machine is allowed to attain it’ssteady state.

4. The buffer level, waste, column count and column temperatureare checked.

5. Theprimer is first run,followed by the blank and finally the calibrator.

6. After the completion of therun, the retention time of HbA2 of the calibrator is

checked.It should be between 3.63-3.67 minutes.

7. If not, then according to the instruction manual, the temperature of the columnis

changed.

8. Only proper calibration allows the samples to run.

9. Each sample takes 6 minutes to run.

Interpretation: (Figure 9)

Based on the level of HbA2, the interpretations are as follows:

HbA2< 3.5 %: Normal

HbA2 3.6-3.9 %: Borderline HbA2[needs confirmation by DNA analysis]

HbA2> 4.0 %: β-thalassemia trait

HbA2< 2.0 %: Can be α-thalassemia / δ-thalassemia heterozygote

Common Hb variants such as HbS, HbDPunjab

andHbQIndia

are identified as separate peaks in

different windows with specific retention times.

Page 43: Training Manual for Hemoglobinopathies and Hemophilia:

43

Page 44: Training Manual for Hemoglobinopathies and Hemophilia:

44

Page 45: Training Manual for Hemoglobinopathies and Hemophilia:

45

Page 46: Training Manual for Hemoglobinopathies and Hemophilia:

46

Figure 9: HPLC chromatograms on Variant II showing different hemoglobinopathies

Note:

Automated HPLC provides a presumptive identification of abnormal Hb variants.

Other methods such as Cellulose acetate electrophoresis at alkaline pH, sickling

test should be used in combination for identification of these variants while final

confirmation of these variants is done by DNA analysis.

Careful examination of the chromatograms is important to identify some structural

Hb variants such as δ-globin gene variants and HbLepore.

Page 47: Training Manual for Hemoglobinopathies and Hemophilia:

47

References:

1. Bain BJ, Lewis SM, Bates I. Basic haematological techiniques. In: Lewis SM, Bain

BJ, Bates I. Dacie and Lewis Practical Haematology, 10th

edition, Churchill

Livingstone p-25 (2006).

2. Itano HA and Pauling L. A rapid diagnostic test for sickle cell anemia. Blood

1949;4:66.

3. Huntsman R.G., Barclay G.P.T., Canning D.M. and Yawson G.I. A rapid whole blood

solubility test to diffrfentiate the sickle cell trait form sickle cell anemia. J. Clin.

Path.1970;23:781.

4. Gorakshakar AC, Colah R, Nadkarni A, Desai S: Evaluation of the single tube

osmotic fragility test in detection of beta thalassemia trait. Natl Med J India

1990;3:171.

5. Mohanty D, Colah R. Eds Laboratory Manual for Screening, Diagnosis and Molecular

analysis in Hemoglobinopathies and Red Cell Enzymopathies 1st Edition. Bhalani

Publishing House, Mumbai 2008.

6. Colah RB, Surve R, Sawant P, D’Souza E, Phanasgaonkar S, Nadkarni AH,

Gorakshakar AC. HPLC studies in hemoglobinopathies. Ind. J. Pediat. 2007;74:31.

Limitations:

HbLepore, HbE and HbD Iran co-elute with HbA2.

HbA2is falsely increased in thepresence of HbS.

HbA2 is falsely reduced in thepresence of HbD.

HbH and Hb Bart’s show a sharp peak at the start of the chromatogram but are not

identified accurately. They must be confirmed by other methods.

Page 48: Training Manual for Hemoglobinopathies and Hemophilia:

48

Patient information sheet

Thalassemia major

This is a life long blood disease that causes serious anemia, needing blood transfusion. It is an

inherited disease due to a minor blood mutation in both parents who do not show any disease.

This is why testing before pregnancy is very important. Prenatal testing can prevent this

disease.

If your child has thalassemia major then good medical care can prolong life and

decrease complications.

Regular blood transfusions are necessary, Keep hemoglobin more than 9 g/ dl before

the regular blood transfusion. If Hemoglobin goes to a very low level each month it

will result in more complications like big spleen, bone problems, poor growth etc.

It is important to check iron levels by serum ferritin. If this is high it is important to

take iron chelation (iron removal). There are 3 iron chelating medicines. Take the

medicine recommended by your doctor regularly. If iron overload is not treated it will

result in more problems e.g.hormone defects, affect the heart and liver and other

organs.

Special tests for better checking foriron overload like T2 * MRI are required. As the

child gets older different problems like bone problems may occur. If the special tests

are not available at your local centre, discuss with your doctor and you can be sent to

a higher centre for these tests and checkup.

Regular monitoring is very important for health, your doctor will give you full

information.

The only cure is Bone marrow transplant, but this may only be possible in some

patients. Also this procedure has risks including a chance of death, this may be higher

in older patients or those with complications. Ask your doctor if this is a suitable

option in your case.

With good blood transfusion and iron chelation medicines you can lead a productive

life.

Page 49: Training Manual for Hemoglobinopathies and Hemophilia:

49

Patient information sheet

Sickle cell disease (SCD)

Sickle cell disease (SCD) is a serious, inherited condition affecting the red blood cells. This causes”

sickling” (shaped like a crescent moon), of the usually round red blood cells, these sickle cells cause

pain and other symptoms and affect many organs of the body.

Sickle cell trait is not the same as sickle cell disease. Sickle cell trait means you carry a sickle cell

gene, but usually such people are not ill.

Diagnosis

We can make a diagnosis by the dried blood spot newborn test which can be confirmed by a

HPLC/Hb Capillary zone electrophoresis blood test. Early diagnosis means that the child will receive

early and appropriate care needed for management.

What problems can be seen?

Pain- mild to severe, pain crisis

Anemia

Jaundice,

Fever ( these children are at risk for serious infections)

Pneumonia etc

Acute chest syndrome

Stroke

Kidney problems etc

Treatment

Good treatment, started early in life, may help to prevent complications. See a specialist about the

disease at least annually.

Take pneumoccalvaccination and oral penicillin medicine to prevent some infections

Some things which can increase sickling, are cold weather, infection, lack of

fluid in the body (dehydration) or low oxygen.

The sickle cells containing mostly HbS are less flexible than normal red

blood cells and get stuck in small blood vessels and block them.

These sickle cells are destroyed more easily than normal red blood

cells. Patients will have a moderate anemia. Some other complications

include pain crisis due to sickling, trapping of blood in spleen,

increased infections, acute chest syndrome etc.

Page 50: Training Manual for Hemoglobinopathies and Hemophilia:

50

Take folic acid and Hydroxyurea as prescribed by the doctor. There is a national programme for

Sickle cell anemia through the government. Visit your state District hospital for details.

Page 51: Training Manual for Hemoglobinopathies and Hemophilia:

51

Newborn Screening for Sickle Cell Disease

Hemoglobinopathies are one of the commonest groups of single gene disorders in the Indian

subcontinent and pose a major drain on our health resources. Sickle cell disease (SCD) is an

important public health problem in Indiawith highest prevalence amongst the tribal and some

non-tribal ethnic groups. Sickle cell disease in India has a very varied clinical presentation

ranging from a severe to mild or asymptomatic condition. Early diagnosis and providing

comprehensive care are critical in SCD because of the possibility of lethal complications in

early infancy in pre-symptomatic children. Children with SCD have an increased

susceptibility to severe bacterial infection, particularly due to Streptococcus pneumoniae

which can occur as early as 4 months of age and carries a case fatality rate as high as 30%.

Acute splenic sequestration crisis also contributes to mortality in infancy. It has been

documented in different countries of the world that pneumococcal sepsis is a major cause of

early mortality in sickle cell disease and prophylactic penicillin therapy provided can

dramatically reduce the morbidity and mortality. Early introduction of penicillin can lead to a

dramatic decrease in early childhood mortality from pneumococcal sepsis. Newbornscreening(NBS) and comprehensive care have dramatically decreased morbidity and

improved survival of patients with SCD.Since the implementation of NBS in developed

countries, there has been a dramatic improvement in the survival of infants with sickle cell

disease.The aim of aNBS program for SCD is to prevent major

healthcomplicationsofSCDfrom early childhood onward. The approaches for a NBS program

could be “Targeted screening” which takes the ethnic ancestry of every newborn into account

or “universal screening” wherethe entire newborn population is screened irrespective of

family origins. Laboratory methods to be usedfor the detection of disease and carrier states should be very

sensitive and highly specific so that missing or falsely identifying patients will be minimized.

Technologies are based on the separation of hemoglobin (Hb) and the quantification of

respective hemoglobin fractions either from fresh cord blood samples or adried blood spot

(DBS) (Figure 10). Storage of cord blood shouldnot exceed seven days at 4°C and that of

dried blood spots should not exceed 1 month.Older. samples show lower elution of samples

from the DBS and, often, the degradation of hemoglobin,resulting in a high baseline,

increased noise, and unclear peaks with. difficulties in zone adjustmentand quantification,

especially in Hb species present at low percentage.

Page 52: Training Manual for Hemoglobinopathies and Hemophilia:

52

The commonly used technologies for NBS for SCD include high performance liquid

chromatography (HPLC), isoelectric focusing (IEF) and capillary electrophoresis (IEF).

Globally, isoelectric focusing (IEF) using eluates from dried blood spots was initially used

for screening of newborn babies for sickle cell disease but at many centres this had been

replaced by HPLC analysis.The Variant NBS machine. (BioRad laboratories) has been used

for hemoglobin analysis from dried blood spots or the Variant Hemoglobin Testing

System(BioRad laboratories) for cord blood samples using either the sickle cell short or the

thal short programmes. The thal short programme had the advantage of picking up other

hemoglobin abnormalities including some rare non deletional chain variants like Hb

Fontainebleau,Hb O Indonesia and Hb Koya Dora.

Figure 10: Newborn screening strategyfor sickle cell disorders

Specimens:

Anticoagulated (EDTA or Heparin) cord blood sample of newborn at birth

Heel prick sample on filter paper

Sample collection:

Cord bloodsample:

Page 53: Training Manual for Hemoglobinopathies and Hemophilia:

53

The blood sample is collected from the umbilical cord at the time of delivery making sure

there is no contamination with maternal blood.

Heel prick sample:

The newborn infant’s heel is cleaned with a disinfectant. The heel is allowed to air dry

and a puncture made using a sterile lancet (Figure 11).

The first drop of blood is gently wiped off with sterile cotton.

The newborn baby’s blood is collected from the second drop on filter paper by

touching the printed side of the filter paper to the blood drop.

The quantity of the blood should be quite enough to absorb and moist the paper from

both the sides (blood should not be applied on both the sides).

The filter paper is allowed to dry properly and kept away from direct sunlight and

heat.

One wet filter paper should never be superimposed on another before drying after

blood has been collected.

The dried filter paper cards can then be sent to the respective laboratories at the

earliest.

Samples collected on filter paper are run on another HPLC machine (NBS HPLC) and

can also be used forthe POC device.

Figure 11: Collection of blood sample from newborns by heel prick

Page 54: Training Manual for Hemoglobinopathies and Hemophilia:

54

Point of care (POC) Testing

Recently, several point-of-care devices have been developed for screening which are either

paper based screening protocols or antibody based rapid diagnostic devices based on lateral

flow immunoassay technologies. They are simple to use and relatively inexpensive as they do

not require any specific equipment or even electricity which is often not always available in

remote rural regions. These commercial devices have been validated for newborn screening

for SCD with a clinical accuracy of 97 to 100%. One such kit is based on competitive lateral-

flow immunoassay that uses monoclonal antibodies to detect hemoglobins A, S, and C in a

1.5-μL sample of whole blood(Figure 12 and 13).

Test procedure should be followed as per the manufacturer’s instruction with the kit

Figure 12: Test procedure for lateral flow immunoassay using monoclonal antibodies to

detect Hb A, Hb S and Hb C.

Page 55: Training Manual for Hemoglobinopathies and Hemophilia:

55

Figure 13: Diagnosis based on lateral flow immunoassay using monoclonal antibodies to

detect Hb A, Hb S and Hb C.

High Performance Liquid Chromatography (HPLC)

Automated cation-exchange HPLC is a widely usedmethod to screen for hemoglobinopathies

with a clear separation and quantification of hemoglobin fractions. Eluted hemoglobin is

detected by adual-wavelength detector and quantified by integrating the area underthe curve

of the producedchromatogram, expressed as a percentage of the total area. Newborn

screening for sickle cell disease can be doneby high performance liquid chromatography

(HPLC). The blood sample is collected either from the umbilical cord at birth or by heel

prick from the newborn baby. Unaffected infants will have largely fetalhemoglobin (HbF)

and some adult hemoglobin (HbA). HPLC has been shown to be effective in detecting

hemoglobinopathies characterized by synthesis of an abnormal hemoglobin molecule

immediately after birth. A baby testing positive for a form of sickle cell disease will have Hb

F with Hb S whereas a sickle cell carrier baby will have HbF, HbA and HbS.

Newborn screening using cord blood analysis on CE-HPLC

Sample preparation: 5μl of cord blood is added to 1 mL of hemolysing solution and

mixedproperly by inverting the tubes and run on the HPLC machine.

Sample processing:

Refer Hb pattern analysis by automated HPLC Variant II, which is described in earlier.

Note: There should not be any maternal blood contamination in the cord blood.

Page 56: Training Manual for Hemoglobinopathies and Hemophilia:

56

Interpretation:

Figure 14: Chromatograms of cord blood samples run on the Variant II machine

Newborn screening from blood sample collected byheel prick on filter paper

Sample Preparation for HPLC (Variant NBS)

1. The dried blood collection cards should be stored at 2 –8°C. The eluates obtained

from the dried blood spots are stable for 48 hours when stored at 2 –8°C, or 24 hours

when stored at 15 – 30°C.

2. One 1/8” diameter disc is punched out using the DBS puncher for each dried blood

spot specimen into a well on the microwell plate.

3. 250 μl deionized water (DIH2O) is added to each well. The microwell plate is left for

elution of the hemoglobin on the bench at room temperature for 10 minutes.

4. Themicrowellplate is gently rotated on a microplate shaker at 500 rpm/min for 5 min

(this is only a guideline)

5. Each microplate is visually inspected to ensure all wells are thoroughly mixed; if

necessary, the plates are rotated for a longer time or at a higher speed.

6. The samples are now ready for analysis.

Page 57: Training Manual for Hemoglobinopathies and Hemophilia:

57

Interpretation:

Figure 15: Chromatograms of newborn blood samples collected on DBS run on Variant

NBS machine

Page 58: Training Manual for Hemoglobinopathies and Hemophilia:

58

Capillary electrophoresis (CE)

Capillary electrophoresis (CE) is a separation technique that separates molecules in an

electric field according to size and charge. CE is performed in a small glass tube called a

capillary that is filled with an electrolyte solution. CE combines two principles of separation

of hemoglobins, theelectrophoretic mobility in alkaline buffer and the electro-osmotic flow

resulting in excellentseparation. Detected hemoglobin fractions can be relativelyquantified

and produce an electropherogram. CE is able to detect and relatively quantify many Hb

variants including and chain abnormalities.

Hemoglobin analysis by CE

Capillary's Neonat Hb kit enables the efficient separation of hemoglobin fractions and

detection of a large number of hemoglobin variants and thalassemiaspatterns.When using

fresh cord blood, samples are prepared by mixing 5 μL of blood with 5 μL of hemolysis

solution. When using the dry blood spots, punched paper disks (3.8 mm) are obtained using

the DBS puncher from the dried blood spot. The disks are placed into the wells of a

microplate containing 50 μL of hemolysis solution and kept into a humid chamber for 2 hrs.

The samples are now ready for analysis.

Interpretation:

Figure 16: Electropherograms of capillary electrophoresis: A) Normal, B) HbS Trait, C) HbS

Homozygous (adopted from Giordano 2012)

Page 59: Training Manual for Hemoglobinopathies and Hemophilia:

59

Isoelectric focusing (IEF)

IEF is a very sensitive method and is widely used at relatively low costs. IEF

separateshemoglobin species according to their isoelectric point on a gel medium with very

high resolution.Hemoglobin variants migrate in a pH gradient to the point where their net

charge becomes zero. Bands arenarrow compared to classical electrophoresis and give a

precise picture. HbF and HbA, aswell as relevant hemoglobins can be separated.

5 l of whole blood is placed in the wells of a microtitration plate containing 0.1 mL of

0.05% KCN. The samples are transferredwith a multiple syringe to a line of Whatman filter

paper placed on a strip of cellophane tape. The tape bearing the samples is then placed on the

cathodal side of the gel.The gel is run for around 2 hours using a cooling system at 40C with

1400 volts. The gel is then fixed in TCA for 15 minutes, stained for 10 minutes in

bromophenol blue and destained overnight till the background is clear. The gel can be either

dried on the plate itself or transferred to a Whatman no. 3 filter paper and dried for

preservation.

Interpretation:

AS SS AA

Figure 17: Isoelectric focusing gel picture (adapted from Claudia F, 2018)

References

1. Colah R, Mukherjee M, Ghosh K. Sickle cell disease in India. CurrOpinHematol.

2014;21:215-23.

2. Colombatti R, Montanaro M, Guasti F, Rampazzo P, Meneghetti G, Giordan M,

Basso G, Sainati L. Comprehensive care for sickle cell disease immigrant patients: a

reproducible model achieving high adherence to minimum standards of care.Pediatr

Blood Cancer. 2012; 59:1275-9.

3. Upadhye D, Das R, Ray J, Acharjee S, Ghosh K, Colah R, Mukherjee M. Newborn

screening for hemoglobinopathies and red cell enzymopathies in Tripura state: A

malaria endemic state in Northeast India. Hemoglobin 2018; ;42:43-46.

Page 60: Training Manual for Hemoglobinopathies and Hemophilia:

60

4. Upadhye DS, Jain D, Nair SB, Nadkarni AH, Ghosh K, Colah RB. First case of Hb

Fontainebleau with sickle haemoglobin and other non-deletional α gene variants

identified in neonates during newborn screening for sickle cell disorders.J.ClinPathol.

2012;65:654-9.

5. Eastman JW, Wong R, Liao CL, Morales DR. Automated HPLC screening of

newborns for sickle cell anemia and other hemoglobinopathies. Clin. Chem. 1996, 42,

704–710.

6. Upadhye DS, Jain DL, Trivedi YL, Nadkarni AH, Ghosh K, Colah RB. Newborn

screening for haemoglobinopathies by high performance liquid chromatography

(HPLC): Diagnostic utility of different approaches in resource-poor settings. Clin.

Chem. Lab. Med. 2014, 52, 1791–1796.

7. Giordano PC. Newborn screening for hemoglobinopathies using capillary

electrophoresis.Methods Mol Biol.2013;919:131-45.

8. Claudia F. Newborn Screening for Sickle Cell Disease and Other

Hemoglobinopathies: A Short Review on Classical Laboratory Methods—Isoelectric

Focusing, HPLC, and Capillary Electrophoresis. Int. J. Neonatal Screen.2018, 4, 39;

doi:10.3390/ijns4040039.

9. Steele C, SinskiA,AsibeyJ,et al. Point-of-care screening for sickle cell disease in low-

resource settings: A multi-center evaluation of HemoTypeSC, a novel rapid test. Am J

Hematol2019;94, 39-45.

Page 61: Training Manual for Hemoglobinopathies and Hemophilia:

61

Molecular Analysis of Hemoglobinopathies

β-thalassemiasarecaused by mutations that result in the reduced output or non-production of

β-globin chains. Depending on the output of the chains, they are designated as 0 or

+

thalassemia. The β-thalassemias are a very heterogeneous group of disorderswhere more than

200 mutations including deletional and non-deletional mutations causing -thalassemia (0 or

+) have been reported so far (http://globin.cse.psu.edu).Homozygosity and heterozygosity

for these mutations and their interaction with abnormal hemoglobins like HbS or HbE result

in variable clinical presentation or just a mild and asymptomatic hypochromic microcytic

anemia. The majority of the defects are single nucleotide substitutions, insertions or

deletions. Thus, molecular methods to identify mutations in β-globin genes often help to

understand the clinical heterogeneity of the thalassemia syndrome and may also help in

therapeutic interventions. Molecular characterization of the β-globin gene is important for

prenatal diagnosis to avoid the birth of affected babies.

At present there are various techniques available for detection of β-globin gene mutations.

Most of the procedures are based on amplification of the respective globin genes by PCR and

subsequent analysis of the PCR product. Here we have described few of these techniques.

1. Covalent Reverse Dot Blot Hybridization (CRDB): For detection of 6 common β-

globin gene mutations and two Hb variants (Hb S and HbE).

2. Gap PCR: For detection of deletional mutation (619 bp deletion).

3. Amplification Refrectory Mutation System (ARMS):Fordetection of common and

uncommon β-globin gene mutations.

4. Restriction Fragment Length Polymorphism –PCR (RFLP-PCR): For detection of Hb

Variants (Hb S and HbDPunjab

).

5. Direct DNA Sequencing (Sanger’s Sequencing): For detection of rare β-globin gene

mutations.

DNA extraction:

From blood using Phenol-chloroform method

Among the most convenient sources of DNA, leucocytes are considered best for the

extraction of human genomic DNA. It is estimated that 10 mL of whole blood yields about

Page 62: Training Manual for Hemoglobinopathies and Hemophilia:

62

250 μg of DNA. The phenol chloroform method was used for the extraction of DNA from the

peripheral venous blood.

Principle:

A phenol-chloroform extraction is a liquid-liquid extraction method, which separates

mixtures of molecules based on the differential solubilities of the individual molecules in two

different immiscible liquids. The RBCs to WBCs ratio is approximately 1000:1/ μl of blood.

So, the RBCs were lyzed by the freeze shock approach. The sample was placed at -200C and

then thawed at RT. The five basic steps for DNA extraction were as follows:

→ Chelation of divalent cations such as Mg2+

and Ca2+

to stop the function of DNase

enzymes and degradation of the DNA

→ Breaking open of the cells to get the intra cellular constituents in solution and remove

membrane lipids by adding a detergent.

→ Removal of cellular and histone proteins bound to the DNA by using a phenol- chloroform

extraction step.

→ Precipitation of the DNA in cold ethanol.

→ Solubilization of the DNA in a slightly alkaline buffer.

The lysis buffer added not only lyses the cells but also has EDTA and NaCl, which chelates

the divalent cations which inhibit the nuclease activity and shield the negative phosphate ends

of DNA that allow precipitation in ethanol respectively.

This was followed by addition is a biological detergent Sodium Dodecyl Sulphate (SDS), that

breaks down cell membranes by dissolving the lipids and proteins of the cell and disrupting

the bonds that hold the membrane together. The phenol: chloroform mixture was then added

in equal volumes to the aqueous DNA to form a biphasic mixture. The proteins precipitate

into the organic phase while the DNA remains in the aqueous phase. Ethanol precipitation is

a method used to concentrate DNA, as it is insoluble in the relatively non polar ethanol while

lipids and proteins are not. This property leads to easy separation of DNA.

Reagents:

Stock solutions:

5 M NaCl: Dissolve 292.5 gm of NaCl in 500 mL of D/W and makethe volumeup to

1000 mL with D/W.

Page 63: Training Manual for Hemoglobinopathies and Hemophilia:

63

0.5 M EDTA (pH 8.0): Dissolve Disodium EDTA -93.06 gm in 200 mL of D/W.

adjust pH to 8.0 with 10 N NaOH and make thevolume up to 500 mL with D/W.

1 M Tris: Dissolve Tris- 121.0 gm in 500 mL of D/W. Adjust pH to 8.0 with

concentrated HCl and make volume up to 1000 mL with D/W.

10 N NaOH:Dissolve NaOH 400 gm in 1000 mL of D/W

20 X SSC (pH 7.0):Dissolve NaCl- 175.3 gm and Sodium Citrate- 88.2 gm in 500 mL

of D/W. Adjust pH to 7.0 with 10 N NaOH and make volume up to 1000 mL with D/W.

10 % SDS:Dissolve 10 gm SDS in 100 mL D/W.

Working solution:

2X lysis buffer: 200 mM NaCl+ 50 mM EDTA

40 mL of 5 M NaCl+ 100 mL of 0.5 M EDTA. Make volume up to 1 litre with D/W.

1X lysis buffer: 100 mM NaCl + 25 mM EDTA

20 mL of 5 M NaCl + 50 mL of 0.5 M EDTA. Make volume up to 1 litre with D/W.

10 % SDS

Proteinase K: 20 mg/ml D/W (It was stored at -200C in aliquots).

Tris equilibrated phenol (pH 8.0)

Melt 100 mL of the distilled phenol at 680C in a water bath and add 8-hydroxy quinoline

to a final concentration of 0.1 %. First saturate phenol by adding an equal volume of

D/W and mix. After the phases separate on standing, aspirate the upper aqueous phase

and equilibrate the phenol with an equal volume of 1 M Tris (pH 8.0). Aspirate upper

aqueous layer and re-equiibrate phenol with 0.1 M Tris (pH 8.0). The pH of the phenolic

phase should be more than 7.8. Store this equilibrated phenol under 0.1 M Tris (pH 8.0)

in a dark bottle at 40C.

Chloroform: Octanol: Mix 24 mL of Chloroform with 1 mL of Octanol

Chilled ethanol

Tris- EDTA (TE) buffer (pH 7.4): 10 mM Tris + 1mM EDTA

1 mL of 1 M Tris (pH 7.5) + 0.2 mL of 0.5 M EDTA (pH 8.0).Adjustthe pH to 7.4 with

HCL and make the final volume up to 100 mL with D/W.

Procedure:

1. Keep 5-8 mL of EDTA blood sample at -200C overnight before the DNA extraction.

2. Allow the samples to thaw at RT for about 30 minutes to 1 hour.

Page 64: Training Manual for Hemoglobinopathies and Hemophilia:

64

3. Transfer blood samples to 50 mL polypropylene tubes and add 40 mL of 2X lysis buffer

and mix well the tubes.

4. Centrifuge tubes for 15 mins at 4000 rpm at 40C.

5. Drain off the supernatant by inverting the tubes and the WBC pellet retained.

6. Add 1.5 mL of 1X lysis buffer to the pellet and mix by vortexing.

7. Add 150 μl of 10 % SDS and 10 μl proteinase K and mix the tubes by vortexing.

8. Incubate samples overnight at 370C in a water bath.

9. Next day centrifuge the tubes at 4000 rpm for 2 minutes and transfer to 7 mL of plastic

tubes.

10. Add an equal volume of phenol to the sample; mix the tubes well by inversion and

centrifuge at 4000 rpm for 15 minutes.

11. Carefully transfer the upper aqueous layer into a new plastic tube. Repeat Step 10.

12. Following centrifugation again transfer the upper aqueous layer to a new plastic tube and

add anequal volume of Chloroform: Octanol.

13. Mix the tubes well by inversion and centrifuge at 4000 rpm for 5 minutes.

14. Take out the upper aqueous layer in another fresh tube.

15. To this add about 5 mL of chilled ethanol and mix by inversion.

16. Lift out the precipitated DNA fibre with a closed Pasteur pipette, air dry and dissolve in

200 to 500 μl of TE buffer (pH 7.4).

17. The quality of the DNA is checked as follows:

Checking for high molecular weight DNA:

Stock 50X TAE buffer: 242 g of Tris base + 100 mL of 0.5 M EDTA + 57.1 mL of

Glacial Acetic acid. Make thevolume up to 1000 mL with D/W.

1X TAE buffer (500 ml):Dilute 10 mL of 50X TAE buffer to 500 mL with D/W

Load 5 μl of DNA with 1 μl of loading dye on a 0.8% agarose gel with ethidium bromide

and run for 1 hour in 1X TAE buffer at 90 volts in a submergedgel electrophoresis tank.

Observe the gel under an ultraviolet transilluminator (UV). A single band near the well

fluoresces which is the DNA. When a smear is observed instead of the band it indicates

that the sample is degraded.

Page 65: Training Manual for Hemoglobinopathies and Hemophilia:

65

Quantification of the high molecular weight DNA:

The DNA concentration and purity is checked by diluting 5μl of DNA with 2ml of distilled

water and reading the O.D. at 260 and 280nm in a spectrophotometer.

1 OD AT 260 nm = 50 μg/ml of DNA

Concentration of DNA (50 μg/ml) = OD at 260 x 50 x dilution factor

OD ratio of 260 nm/280 nm should be between 1.6-1.8. If the ratio is less than 1.6, it

indicates protein contamination.

Polymerase Chain Reaction (PCR)

PCR is a technique used in molecular biology to amplify a region of DNA of interest to

generate thousands to millions of copies.

Principle:

It is based on the principle of thermal cycling which consists of repeated heating and cooling

of the reaction for DNA melting and enzymatic replication of DNA. Short DNA fragments

(primers) containing sequences complementary to the target region along with DNA

polymerase carry out the amplification process. The thermal cycling process physically

separates the two strands of the DNA double helix and then each strand is used as a template

to selectively amplify the region of interest. Thus, it is a chain reaction in which the DNA

template is exponentially amplified.

Components and reagents required:

DNA template: The region of interest to be amplified.

Primer set: Forward and Reserve sequences complementary to the 3’ ends of the sense

and antisense strands of the target DNA respectively.

Taq DNA polymerase: The enzymatic replication of DNA (the source of the enzyme is

the microorganism Thermusaquaticus).

Page 66: Training Manual for Hemoglobinopathies and Hemophilia:

66

Deoxyribonucleotide triphosphates (dNTPs): There are 4 dNTPs (dA,dT, dG, dC)

required for extension. These are building blocks from which the DNA polymerase

synthesizes the new strands.

Divalent cation generally Mg+2

: MgCl2

Buffer:Provides a suitable chemical environment for optimum activity and stability of

DNA polymerase.

PCR involves 3 main steps: (Figure 18)

Denaturation: This step is carried out at 940C- 98

0C for 20-30 sec. It causes the

denaturation of the DNA template by disrupting the hydrogen bonds between

complementary bases yielding separation of two strands of the DNA into two single

strands of DNA.

Annealing: In this step the temperature is lowered to 50-650C for 20-40 seconds. This

allows the annealing of the primers to the single stranded DNA template. The Taq DNA

polymerase binds to the primer-template hybrid and initiates DNA synthesis in the next

step.

Extension: The temperature is usually 720C for this step. The DNA polymerase

synthesizes a new DNA strand complementary to the DNA template by adding dNTPs in

the 5’ 3’ direction condensing the 5’ phosphate group of the dNTPs with the 3’

hydroxyl group at the end of the extending strand.

This cycle of 3 steps is repeated 30-35 times.

Final extension: This step is performed at a temperature of 720C (used in the extension step)

for 5-15 mins. This step ensures that any remaining single stranded DNA is fully extended.

Page 67: Training Manual for Hemoglobinopathies and Hemophilia:

67

Figure 18: Steps involved in PCR.

Mutation detection

I. Detection of the six common Indian β- thalassemia mutations and two

hemoglobinvariants, Hb S and Hb E by Covalent Reverse Dot Blot (CRDB)

Hybridization.

Summary and explanation of the test:

β- thalassemia is one of the commonest single gene disorders and is caused mainly by point

mutations in the β- globin gene. Six common Indian mutations namely IVS 1 nt 5 (G>C),

IVS 1 nt 1 (G>T), CD 8/9 (+G), CD 41/42 (-CTTT), CD 15 (G>A) and CD 30 (G>C) along

with two structural variants of haemoglobin, HbS and HbE are screened for by hybridization

with allele specific oligonucleotide probes followed by colour development. Using this

method we can distinguish between heterozygous, homozygous and compound heterozygous

states of β- thalassemia and these abnormal hemoglobins (Figure 19).

Reverse dot blot analysis was first described by Saiki et al in 1989 and then developed later

to screen many β-thalassemia mutations and for use in prenatal diagnosis. This method can

Page 68: Training Manual for Hemoglobinopathies and Hemophilia:

68

also be used for confirmation of the diagnosis of the cases identified during newborn

screening.

Figure 19: Schematic representation of the principle of Covalent Reverse Dot Blot

Hybridization (CRDB)

Interpretation:

The mutations are identified on the basis of the spots obtained on the membrane by the

biotin- streptavidin- alkaline phospathase reaction(Figure 20 and 21).

Figure 20: Schematic representation of the preparation of the oligoprobes blotted

membrane.

Page 69: Training Manual for Hemoglobinopathies and Hemophilia:

69

Figure 21: Covalent Reverse Dot Blot Hybridization showing different β-globin gene

mutations.

II. Detection of the 619 bp deletion causing β-thalassemia

The 619 bp deletion is another common β globin gene mutation in India. It is a β0 type of

mutation. It spans from exon 2 to IVS II of the β globin gene.

Principle:

A direct PCR and electrophoresis is used to detect this mutation. Two primers are used

named as 2a and 2b which amplify a 861 bp fragment from the 3’ end of the - globin gene.

Each primer lies on either side of the 619 bp deletion, therefore they directly detect this

mutation by amplifying a characteristic smaller fragment of 242 bp.

Reagents:

Primers: (Sigma Genosys, India)

Table 3: Primers used to amplify theregion of the -globin gene

Primers Sequence

2a 5’ CAA TGT ATC ATG CCT CTT TGC ACC 3’

2b 5’ GAG TCA AGG CTG AGA GAT GCA GGA 3’

Note: This method can be used as a second alternative when commercial kits become

available

Page 70: Training Manual for Hemoglobinopathies and Hemophilia:

70

dNTP mix (25mM)

PCR buffer

Taq Polymerase

Distilled water

Genomic DNA

PCR Reaction:

Table 4: Reagents used to detect the619 bp deletion

Reagents Concentration Volume

DNA 100-200 ng/ µl 2 µl

2a 10 picomoles/ µl 2 µl

2b 10 picomoles/ µl 2 µl

dNTP mix 25 mM 0.5μl

PCR buffer 10X containing 15mM MgCl2) 5.0μl

MgCl2 25mM 7.0μl

Taq Polymerase 5 units/ µl 0.25 μl

Distilled water -- 30.0μl

Total 50 μl

Procedure:

1. Set up PCR reactionin a single tube.

2. Add DNA

3. Vortex the tubes containing premix after addition of Taq polymerase and centrifuge. Place

the tubes in the PCR machine.

PCR Programme:

Table 5: PCR program to detect the619 bp deletion

Denaturation 940C for 5 mins

Annealing 550C for 3 mins

35 cycles of

Extension 720C for 2 min

Denaturation 940C for 1 min

Annealing 550C for 1 min

Final extension 720C for 7 mins

Hold 150C for

Check the result in 2% agarose gel.

Page 71: Training Manual for Hemoglobinopathies and Hemophilia:

71

Reagents

1X TAE Buffer

Gel loading dye

2% agarose gel

Procedure:

1. Add 2 µl of loading dye to the tube containing the PCR product.

2. Load the entire product on a 2% agarose gel.

3. Load the molecular weight marker VIII (Roche) side by side to check for specific

amplification.

4. Run the gel at 80-100 V for 1 hour and view under UV light on a gel documentation

system and record the results.

Interpretation:

Agarose gel electrophoresis

Figure 22: Schematic representation of detection of the 619 bp deletion mutation by

GAP- PCR.

Lane 1: DNA molecular weight marker

Lane 2: 619 bp del heterozygous

Lane 3: 619 bp del homozygous

Lane 4: Normal

Page 72: Training Manual for Hemoglobinopathies and Hemophilia:

72

Figure 23: 2% agarose gel electrophoresis showing the619 bp deletion.

III. Detection of β thalassemia mutations by Amplification Refractory Mutation System

(ARMS).

The amplification-refractory mutation system (ARMS) technique for detecting known point

mutations was first described by Newton et al in 1989. This method is also used to detect

known thalassemia mutations present among the Indian population.

Principle:

The technique is based on the principle of allele-specific priming of the PCR process. Allele

specific primers are used such that the nucleotide at the 3’ end of each primer is

complementary to the change of DNA sequence caused by the mutation that is being

screened. To enhance their specificity, a deliberate additional mismatch is introduced at

position - 4 from the 3’ end. Thus for every sample, two reactions are set up, one containing

the normal primer and the other containing the mutant primer. Along with these primers

another set of primers are also added which span a completely different part of the gene and

this serves as a control fragment thus confirming the amplification of the sample and that the

PCR has worked. Each of these primers is run along with the positive and negative control

DNA samples under uniform stringent PCR conditions to ensure that successful amplification

occurs only in the presence of the mutation (Figure 24).

Lane 1: DNA molecular weight marker

Lane 2: 619 bpdel heterozygous

Lane 3: 619 bpdel homozygous

Lane 4: Normal

Page 73: Training Manual for Hemoglobinopathies and Hemophilia:

73

Figure 24: Diagrammatic representation of principle of ARMS PCR.

Reagents:

dNTP stock Solutions (1.25mM)

60 μldATP (100 mM) + 60 µl dTTP (100 mM) + 60 µl dCTP (100 mM) + 60 µl dGTP

(100 mM) + 4760 µl of sterile D/W. Store at -200C.

ARMS stock Master Mix: Aliquots of this mix can be preserve at -20 0C

Table 6: Reagents required to prepare the mastermix for ARMS PCR

Reagents Volume

10X PCR Buffer containing 15 mM MgCl2 1000 µl

Sterile D/W 5400 µl

dNTP mix (1.25 mM) 1600 µl

Spermidin (1 M solution) Sigma 8 µl

Table 7: Primers used to detect different -globin gene mutations by ARMS

Fragment Primers Size (bp) Sequence (5’3’)

Set I

Control 2a 860 CAA TGT ATC ATG CCT CTT TGC ACC

2b GAG TCA AGG CTG AGA GAT GCA GGA

Common (Forward) CM 23 ACC TCA CCC TGT GGA GCC AC

Normal and Mutant

(Reverse)

IVS 1-1 (G>A) 281 N: TTA AAC CTG TCT TGT AAC CTT GAT ACC CAC

281 M: TTA AAC CTG TCT TGT AAC CTT GAT ACC GAT

Cd 16 (-C) 239 N: TCA CCA CCA ACT TCA TCC ACG TTC ACG TTG

238 M: TCA CCA CCA ACT TCA TCC ACG TTC ACG TTC

Cd 5 (-CT) 204 N: ACA GGG CAG TAA CGG CAG ACT TCT CCG CAG

202 M: ACA GGG CAG TAA CGG CAG ACT TCT CCG CGA

Cd 30 (G>A) 280 N: TAA ACC TGT CTT GTA ACC TTG ATA CCT ACC

280 M: TAA ACC TGT CTT GTA ACC TTG ATA CCT ACT

Cd 39 (C>T) 436 M: CAG ATC CCC AAA GGA CTC AAA GAA CCT GTA

IVS 1-5 (G>C) 285 N: CTC CTT AAA CCT GTC TTG TAA CCT TGT TAC

285 M: CTC CTT AAA CCT GTC TTG TAA CCT TGT TAG

IVS 1-1 (G>T) 281 M: TTA AAC CTG TCT TGT AAC CTT GAT ACG AAA

Cd 41/42

(-CTTT)

443 N: GAG TGG ACA GAT CCC CAA AGG ACT CAA AGA

439 M: GAG TGG ACA GAT CCC CAA AGG ACT CAA CCT

Cd 8/9 (+G) 214 N: CCT TGC CCC ACA GGG CAG TAA CGG CAC ACT

Page 74: Training Manual for Hemoglobinopathies and Hemophilia:

74

215 M: CCT TGC CCC ACA GGG CAG TAA CGG CAC ACC

Cd 6 (AT)

HbS

207 N: CCC ACA GGG CAG TAA CGG CAG ACT TCT GCT

207 M: CCC ACA GGG CAG TAA CGG CAG ACT TCT GCA

Cd 26 (G>A)

HbE

236 N: TAA CCT TGA TAC CAA CCT GCC CAG GGC GTC

236 M: TAA CCT TGA TAC CAA CCT GCC CAG GGC GTT

Cd 30 (G>C) 280 N: TAA ACC TGT CTT GTA ACC TTG ATA CCT ACC

280 M: TAA ACC TGT CTT GTA ACC TTG ATA CCT ACG

IVS 1-(3’End)-

25 bp Deletion

386 N: GCA GCC TAA GGG TGG GAA AAT AGA CCA ATA

386 M: CTC TGG GTC CAA GGG TAG ACC ACC AGC ATA

IVS II-I (G>A) 634 N: AAG AAA ACA TCA AGG GTC CCA TAG ACT GAC

634 M: AAG AAA ACA TCA AGG GTC CCA TAG ACT GAT

Set II

Control 2a 860 CAA TGT ATC ATG CCT CTT TGC ACC

2b GAG TCA AGG CTG AGA GAT GCA GGA

Common (Reverse) CM 24 CCC CTT CCT ATG ACA TGA ACT TAA

Normal and Mutant

(Forward)

Cap site +1

(A>C)

582 N: ATA AGT CAG GGC AGA GCC ATC TAT TGG TTA

582 M: ATA AGT CAG GGC AGA GCC ATC TAT TGG TTC

-88 (C>T) 684 N: TCA CTT AGA CCT CAC CCT GTG GAG CCA CTC

684 M: TCA CTT AGA CCT CAC CCT GTG GAG CCT CAT

Cd 15 (G>A) 500 N: TGA GGA GAA GTC TGC CGT TAC TGC CCA GTG

500 M: TGA GGA GAA GTC TGC CGT TAC TGC CCA GTA

IVS 1-1 (G>T) 454 N: GAT GAA GTT GGT GGT GAG GCC CTG GGT AGG

Cd 39 (C>T) 299 N: TTA GGC TGC TGG TGG TCT ACC CTT GGT CCC

Set III

Control Hind III A 326 AGT GCT GCA AGA AGA ACA ACT ACC

Hind III B CTC TGC ATC ATG GGC AGT GAG CTC

Common (Reverse) 2b GAG TCA AGG CTG AGA GAT GCA GGA

Normal and Mutant

(Forward)

IVS II -837

(T>G)

646 N: CCT TTT GCT AAT CAT GTT CAT ACC TGC TAT

646 M: CCT TTT GCT AAT CAT GTT CAT ACC TGC TAG

Poly A (T>C) 380 N: TGA GCA TCT GGA TTC TGC CTA AT

380 M: TGA GCA TCT GGA TTC TGC CTA AC

IVS II -745

(C>G)

738 N: TCA TAT TGC TAA TAG CAG CTA CAA TCG AGC

738 M: TCA TAT TGC TAA TAG CAG CTA CAA TCG AGG

IVS II -654

(G>T)

829 N: GAA TAA CAG TGA TAA TTT CTG GGT TAA CGC

829 M: GAA TAA CAG TGA TAA TTT CTG GGT TAT CTG

PCR Reaction:

Table 8: Reagents to amplify the normal and mutant gene using ARMS technique

Procedure:

1. Set up the PCR reactionin two tubes for each DNA sample to detect a single mutation.

Page 75: Training Manual for Hemoglobinopathies and Hemophilia:

75

2. Label them as normal (N) and mutant (M).

3. Add DNA.

4. Add a pair of control primers in each tube to see whether the PCR reaction is working.

5. Vortex the tubes containing premix after addition of Taq polymerase and centrifuge. Place

the tubes in the PCR machine.

PCR Programme:Table 9: PCR program for ARMS PCR

Denaturation 940C for 5 mins

25 cycles of

Denaturation 940C for 1 min

Annealing 650C for 1 min

Extension 720C for 1 min 30 sec

Final extension 720C for 3 mins

Hold 150C for

Check the result in 2% agarose gel.

Reagents

1X TAE Buffre

Gel loading dye

2% agarose gel

Procedure:

1. Add 2 µl of loading dye to the tube containing the PCR product.

2. Load the entire product on a 2% agarose gel.

3. Load the molecular weight marker VIII (Roche) side by side to check for specific

amplification.

4. Run the at 80-100 V for 1 hour and view under UV light on a gel documentation system

and record the results.

Interpretation:

Page 76: Training Manual for Hemoglobinopathies and Hemophilia:

76

Figure 25: Schematic representation of detection of mutations by ARMS

Figure 26: 2% agarose gel showing ARMS PCR for detection of Hb S

1: Molecular weight marker

2: Normal

3: Heterozygous

4: Homozygous

1: Molecular weight marker

2: Normal

3: HbSHeterozygous

4: HbSHomozygous

ARMS PCR will be used as the method of choice for detection of mutations causing

thalassemias and abnormal hemoglobins

Page 77: Training Manual for Hemoglobinopathies and Hemophilia:

77

III. Detection of Hemoglobin S and HemoglobinHbDPunjab

by Restriction Enzyme

Digestion: The main use of this PCR technique is for the diagnosis of the clinically important

Hb Variants Hb S and Hb DPunjab

.

Principle:

A small number of the β-thalassemia mutations create or abolish a restriction endonuclease

recognition site in the globin gene sequence. Hence direct detection of the mutation is

possible by amplification of a fragment of the gene containing the mutation followed by

restriction enzyme digestion and agarose gel electrophoresis (Old JM 2001).

Reagents:

Table 10: Primers used to amplify a specificregion of the-globin gene

Name of the Hb

Variant

Name of the

primer

Sequence (5’3’)

Hb S Hemat 1 F TGG TAT GGG GCC AAG AGA TA

Hemat 2 R AAC GAT CCT GAG ACT TCC ACA

Hb DPunjab

2a CAA TGT ATC ATG CCT CTT TGC ACC

2b GAG TCA AGG CTG AGA GAT GCA GGA

dNTP mix (25mM)

PCR buffer

Taq Polymerase

Distilled water

Genomic DNA

Restriction Enzymes

Dde 1: For Hb S

Eco RI: For HbDPunjab

PCR reaction:

Table 11: PCR reaction for detection of HbS and HbDPunjab

Reagents Concentration Volume

DNA 100 ng/ µl 2.5 µl

Forward primer: Hemat 1 (For HbS) 2a (For HbD) 10 pmoles/µl 2 µl

Reverse primer: Hemat 2 (For HbS) 2b (For HbD) 10 pmoles/µl 2 µl

dNTP Mix 25 mM 1 µl

PCR Buffer (Complete) 10 X 5 µl

MgCl2 25 mM 7 µl

Taq polymerase 5 u/ µl 0.5 µl

D/W 30 µl

Total 50 µl

Page 78: Training Manual for Hemoglobinopathies and Hemophilia:

78

Procedure:

1. Set up thePCR reaction in a single tube.

2. Add DNA

3. Vortex the tubes containing thepremix after addition of Taq polymerase and

centrifuge. Place the tubes in the PCR machine.

4. Always use known normal, homozygous and heterozygous samples as controls.

Table12: PCR programme for detection of HbS and HbDPunjab

Checking for amplification

Reagents

Stock solution: 50 X TAE buffer: 242 gm of Tris + 100 mL of 0.5 M NaCl + 57.1 mL of

Glacial acetic acid. Make thevolume up to 1 litre with D/W.

Working solution: 1 X

Gel loading dye: 125 mg of Xylene Cyanol+ 125 mg of Bromo phenol blue+ 20 gm of

Sucrose. Make thevolume up to 50 mL with D/W.

1% Agarose: Dissolve1gm of agarose in 100 mL of 1X TAE by boiling in a microwave.

Keep at RT to cool to a temperature of around 35-40 0C. Add 5 µl of Ethidium Bromide

to the molten agar and mix well. Pour the molten gel in a gel casting tray with the well

forming comb in place. Allow the gel to solidify at RT.

Procedure:

1. Mix 5 µl of the PCR product with 2 µl of gel loading dye in a loading tray.

2. Load 7 µl of total mixture (5 µl PCR product + 2 µl Dye) on 1% agarose gel.

Page 79: Training Manual for Hemoglobinopathies and Hemophilia:

79

3. Also load the molecular weight marker VIII (Roche) side by side to check for specific

amplification.

4. Run the gel at 80-100 V for half an hour and view under UV light on a Gel

Documentation System.

5. The remaining 45 µl of the PCR product will be used for restriction enzyme digestion.

Table 13: Restriction enzyme digestion

Reagents Concentration For HbS For HbDPunjab

Amplified Product -- 20.0 μl 10.0 μl

Buffer 10X 2.0 μl 2.0 μl

Enzyme 10U/μl 1.0 μl 1.0 μl

D/W -- 0.0 μl 7.0 μl

Total -- 23.0 μl 20.0 μl

1. Mix and centrifuge (pulse spin) the reaction mixture

2. Digest the product by keeping overnight at 370C

Checking the results in 3% agarose gel (for HbS) and 2% agarose gel (for HbDPunjab

)

Reagents

1X TAE buffer

Gel loading dye

3% and 2% agarose gel

Procedure:

1. Add 2 μl of loading dye to the digested products.

2. Load the products on the gel.

3. Load the molecular weight marker VIII (Roche) side by side to check specific band

size.

4. Always run one undigested product with the digested products to check the activity of

theenzyme.

5. Run the gel at 80-100 V for half an hour and view under UV light on a Gel

Documentation System.

Page 80: Training Manual for Hemoglobinopathies and Hemophilia:

80

Interpretation:

Figure 27: 3 % agarose gel electrophoresis after Restriction enzyme digestion using

theenzyme Dde 1.

Figure 28: 2 % agarose gel electrophoresis after Restriction enzyme digestion using the

enzyme Eco R1.

Lane 1: Undigested product

Lane 2: HbS homozygous

Lane 3: Normal

Lane 4: HbS heterozygous

Lane 5: Molecular weight marker

Lane 1: Molecular weight marker

Lane 2: Undigested product

Lane 3: HbDPunjab

heterozygous

Lane 4: HbDPunjab

homozygous

Lane 5: Normal

Note: Presence of HbDPunjab

abolishes the restriction site for Eco R1 and hence

homozygous HbDPunjab

will remain uncut.

Page 81: Training Manual for Hemoglobinopathies and Hemophilia:

81

IV. Automated DNA sequencing:

This technique is based on Sanger’s dideoxy sequencing. DNA sequencing allows the

analysis of the exact nucleotide sequence of the gene or area of a gene, under study. This

method is used to detect other rare or novel-globin gene mutations which remain undetected

by CRDB hybridization and ARMS.

Principle:

The principle of automated DNA sequencing is same as the Sanger's chain termination

method. But unlike Sanger’s method, which uses radioactive material to label the ddNTPs,

cycle sequencing uses fluorescent dyes to label the ddNTPs. The fluorescently labelled

fragments are separated by a high-resolution polymer in a capillary, called POP7

(Performance of polymer) which then pass through a laser beam at the bottom of the gel

where each ddNTP would fluoresce a different color for each of the 4 nucleotides. The laser

excites the fluorescent molecule, which sends out light of a distinct color. This light is

collected and focused by lenses into a spectrograph. Based on the wavelength, the

spectrograph separates the light across a CCD camera (charge coupled device). Each base has

its own color, so the sequencer can detect the order of the bases in the sequenced gene

(Figure 29).

Figure 29: Schematic diagram showing the principle of Sanger's DNA Sequencing

Preparation of the sample:

The target DNA is amplified with a single set of primers (forward and reverse), the target

DNA is run on a 1.0% agarose gel containing ethidium bromide to check for amplification of

Page 82: Training Manual for Hemoglobinopathies and Hemophilia:

82

the PCR product. If there are no non-specific bands, the PCR product can be used directly for

sequencing, or it is purified using gel extraction (Bio Basics Canada INC. EZ-10 spin column

DNA, using the manufacturer’s instructions) or ethanol precipitation.

Amplification of target DNA:

Reagents

Primers:

Table 14: Primers used to amplify the region of interest

Primer Name Sequence (5'3') Size

(bp)

Region of the -

globin gene

amplified

β-fragment 1 F CGA TCT TCA ATA TGC TTA CCA AGC TGT GA 1400 Exon 1, IVS I,

Exon 2 and IVS II β-fragment 1 R TGG TGC AAA GAG GCA TGA TAC ATT GT

2a CAA TGT ATC ATG CCT CTT TGC ACC 860 IVS II and Exon 3

2b GAG TCA AGG CTG AGA GAT GCA GGA

dNTPs

PCR Buffer

MgCl2

TaqPolymarase

Distilled water

Genomic DNA

PCR reaction:

Table 15: Reagents required to amplify thefirst region of interest

Reagents Concentration Volume

DNA 100 ng/ µl 2.5 µl

Forward primer:β-fragment 1F/2a 10 pmoles/µl 2 µl

Reverse primer: β-fragment 1R/2b 10 pmoles/µl 2 µl

dNTP Mix 25 mM 1 µl

PCR Buffer (Complete) 10 X 5 µl

MgCl2 25 mM 7 µl

Taq polymerase 5 u/ µl 0.5 µl

D/W 30 µl

Total 50 µl

Page 83: Training Manual for Hemoglobinopathies and Hemophilia:

83

Note: To amplify β-fragment 1 region use β-fragment 1F and β-fragment 1R primers and to

amplify 2a2b region use 2a and 2b as forward and reverse primers respectively

PCR programme: Table 16: PCR programs to amplify the-globin gene

Gel extraction: (As per the Kit manual)

Procedure:

1. Load the amplified product on 1% agarose gel containing ethidium bromide.

2. Run the gel at 80-100 V for half an hour and view under UV light on a Gel Documentation

system.

3. Excise theDNA fragment of interest from the agarose gel with a clean, sharp scalpel and

transfer to a 1.5 mL tube.

4. Add 400 µl of binding buffer II for each 100 mg of gel to the tube.

5. Incubate a tube containing a mixture of the gel slice and Binding buffer II at 500C-60

0C

for 10 minutes and shake occasionally until the agarose is completely dissolved.

6. Add the above mixture to the EZ-10 column and centrifuge at 10,000 rpm for 2 minutes;

discard the flow-through.

7. Add 750 µl of wash solution to the column and centrifuge at 10,000 rpm for 1 minute;

discard the solution in the tube.

8. Repeat the above step (step 7),centrifugethe tube for 3 minutes at 10,000 rpm to remove

any residual wash solution.

9. Place the column in a clean 1.5 mL tube and add 30-50 µl of elution buffer to the centre of

the column.

Page 84: Training Manual for Hemoglobinopathies and Hemophilia:

84

10. Incubate the column for 15-20 minutes at room temperature and centrifuge at 10,000 rpm

for 3 minutes.

11. Store the purified DNA at -200C.

Cycle sequencing of the desired product:

Reagents:

Purified DNA

Big Dye Terminating sequencing Buffer (ABI 310)

Ready reaction Mix (ABI 310.)

Distilled water

PCR reaction:Table17: Reagents required forcycle sequencing

Reagents Concentration Amount

Purified DNA 100 ng 1-2 µl

Buffer 5X 2

Ready reaction mixture (RR) - 1

Primer (F/R) 3.5 picomole 1

D/W - 14

Total - 20

PCR programme:Table18: PCR program for Cycle sequencing

25 cycles

Initial Denaturation 96° C 10 secs

Annealing 60°C 4 mins

Final Hold 4°C -

Purification of the cycle sequencing product:

1. Add 80 μl of HPLC purified D/W to 20 μl of the product.

2. In a fresh eppendorf (1.5 ml), add 10.0 μl of 3M sodium acetate (pH-4.6) and 250 μl of

absolute ethanol. Add the above reaction mixture to the eppendorf and mix properly by

inverting the tubes.

3. Centrifuge the eppendorf at 14,000 rpm for 20 mins.

4. Discard the supernatant immediately after centrifugation.

Page 85: Training Manual for Hemoglobinopathies and Hemophilia:

85

5. Add 250 μl of 70% ethanol to the pellet, mix gently and centrifuge at 14,000rpm for

15minutes.

6. Discard the supernatant and repeat the above step (step 5).

7. After the above centrifugation, discard the supernatant completely and allow the pellet to

dry.

8. After drying the pellet, add 20 μl of Template suppressor reagent (Hi-DiTMFormamide),

vortex and incubate for 5mins at R.T.

9. Spin down the samples and transfer thecontents into the 96-well plate. Denature the

samples in the plate at 95°C for 5mins and then immediately transfer to ice.

10. Place the plate on the loading tray of the sequencer and subject to sequencing using the

ABI Prism data collection software.

Interpretation:

Figure 30: Identification of 3 rare globin gene mutations by sequencing

Page 86: Training Manual for Hemoglobinopathies and Hemophilia:

86

Referances:

1. Weatherall D, Akinyanju O, Fucharoen S, Olivieri N, Musgrove P. Inherited

Disorders of Hemoglobin. Weatherall D, Akinyanju O, Fucharoen S, Olivieri

N, Musgrove P.Editors In: Jamison DT, Breman JG, Measham AR, Alleyne

G, Claeson M, Evans DB, Jha P, Mills A, Musgrove P, editors. SourceDisease

Control Priorities in Developing Countries. 2nd edition. Washington (DC): World

Bank; 2006. Chapter 34.

2. http://globin.cse.psu.edu

3. Mohanty D, Colah R. Eds Laboratory Manual for Screening, Diagnosis and Molecular

analysis in Hemoglobinopathies and Red Cell Enzymopathies 1st Edition.Bhalani

Publishing House, Mumbai 2008.

4. Colah RB, Gorakshakar AC, Lu Cy, Nadkarni AH, Desai SN, Pawarf AR, Lulla CP,

Krishnamoorthy R and Mohanty D. Application of Covalent Reverse Dot Blot

Hybridization for Rapid prenatal diagnosis of the common Indian thalassemia

syndromes. Indian J Hematol Blood Transfus.1997;15:10.

5. Saiki RK, Walsh PS, Levenson CH, Erlich HA. Genetic analysis of amplified DNA

with immobilized sequence—specific oligonucleotide probes. Prot. Natl Acad Sci

USA. 1989;86: 6230-623.

6. Old JM. DNA based diagnosis of the hemoglobin disorders, In ‘Disorders of

hemoglobin: Genetics, pathophysiology and clinical management. Eds. Steinberg

MH, Forget BG, Higgs DR, Nagal RL, Cambridge University press, Cambridge pp

946-951. 2001.

7. Newton CR, Graham A and Heptinstall LE. Analysis of any point mutations in DNA.

The amplification refractory mutation system (ARMS). Nucl Acids Res 1989;17:

2503-2516

8. Sanger F, Nicfklen S, Carlson AR. DNA sequencing with chain terminating

inhibitors. Proc Natl Acad Sci 1977;74:5463-5467.

Page 87: Training Manual for Hemoglobinopathies and Hemophilia:

87

Prenatal Diagnosis of Hemoglobinopathies

Prenatal diagnosis is a crucial component of a prevention programme for

hemoglobinopathies. It is an important option for high- risk couples where both partners are

carriers of hemoglobinopathy genes to have a normal child. The facilities for prenatal

diagnosis became available in India in mid 1980s. The techniques employed for prenatal

diagnosis include Chorionic Villus sampling (CVS) or Amniocentesis and DNA analysis or

Cordocentesis and fetal blood analysis.

Chorionic villus sampling (CVS)

This technique is used to obtain fetal tissue for prenatal diagnosis of hemoglobinopathies.

The sampling is usually done in the 1st trimester (10.5 -12 weeks of gestation). This

procedure can be done in two ways transcervical and transabdominal. The chances of

spontaneous miscarriage is more with the trancervical route hence transabdominal route is

highly preferable. CVS has an advantage that it is a first trimester procedure and hence

reduces both the emotional stress and the complications associated with 2nd

trimester

diagnosis. Also, CVS provides a good source of fetal DNA which allows quick results in few

days. The risk of fetal loss is minimal.

Amniocentesis:

Amniocentesis is done in the 2nd trimester between 14th

-16th

weeks of gestation. Around 30

to 35 mL of amniotic fluid is needed to get sufficient amount of fetal DNA for prenatal

diagnosis of hemoglobinopathies.

Amniocentesis is associated with 1% fetal loss. Sometimes there is insufficient DNA

obtained from the fluid which may require culturing of the cells and further delaying of the

diagnosis. Minor complications following amniocentesis include continuous leakage of

amniotic fluid, bleeding, and uterine irritability.

Cordocentesis:

The other name for this procedure is umbilical cord blood sampling or fetal blood sampling.

It is done between 18th

-20th

weeks of gestation for second trimester prenatal diagnosis of

hemoglobinopathies. This procedure is only done when couples at risk of having an affected

Page 88: Training Manual for Hemoglobinopathies and Hemophilia:

88

child are identified late or if the diagnosis done during the first trimester is not conclusive.

The fetal blood sample can be analyzed by HPLC.

Figure31: Different methods to obtain fetal samples for prenatal diagnosis.

Despite these advances, shortcomings are there in the invasive techniques leading to 0.5-2%

fetal loss. Hence non-invasive diagnostic procedures are being developed to avoid risk of

miscarriage and to provide sufficient retrieval of fetal DNA for precise diagnosis. These

approaches are based on isolating and analyzingfetal cells in the maternal circulation or

circulatory cell free fetal DNA from maternal plasma.

Page 89: Training Manual for Hemoglobinopathies and Hemophilia:

89

Sample Processing:

Chorionic villus sampling (CVS):

TheCVS sample isusually collected in sterile RPMI medium.

Once the sample is collected, it should be immediately sent to the respective

laboratories for further analysis.

As soon as the laboratory receives the sample, the first step is to clean the

CVS under aninverted dissecting microscope to remove maternal

contamination.

Cleaning of CVS should be done carefully since it can affect the diagnosis of

the fetus.

The cleaned CVStissueis resuspended in CVS Buffer ina 1.5 mL of

eppendorftube.

Preparation of CVS buffer

1.2 mL of 5 M NaCl + 2 mL of 0.5 M EDTA, and make thevolume up

to 40ml with sterile D/W.

Centrifuge the tube at 12000 rpm for 5 mins

Proceed further for DNA extraction

Amniotic fluid (AF):

Centrifuge the tube containing AF at 4000 rpm for 30 mins

Discard the supernatant and add normal saline ( 500μl to 1000μl) to the pelette

of amniocytes

Mix by tapping and transfer the saline with the cells in a1.5 mL eppendorftube

Centrifuge the tube at 12000 rpm for 3 mins

Discard the supernatant

Add 500-1000μl of normal saline to the cells.

Mix by tapping , Centrifuge the tube at 12000 rpm for 3 mins

Proceed further for DNA extraction

Cord blood sample/Fetal blood sample:

Cord blood sample of the fetusis collectedina heparin tube

Complete blood count should be done to know the value of MCV. It should be

>100 fl.

Page 90: Training Manual for Hemoglobinopathies and Hemophilia:

90

Fetal cell staining (Kleihauer –Betke method) is to be done to know the % of F

cells in the blood. There should not be any adult cell in the cord blood.

(Method is described below)

Presence of adult cells in the cord blood indicates that the cord blood is

contaminated with the maternal blood. Contaminated blood cannot be used for

further diagnosis. Repeat sampling has to be done.

The uncontaminated fetal blood is further processed for CE- HPLCto

determine the level of HbF,HbA0 and HbS (incase of Sickle cell) (Method and

interpretation are described below)

Proceed for DNA extraction

Fetal Cell Staining: Kleihauer –Betke Method

Principle: HbF is not uniformly distributed among red cells, except in the condition of

deletional HPFH. Cells with detectable amount of HbF are called F cells and they can be

detected on the blood smear by the Kleihauer-Betke acid elution test. This method is based on

elution of adult hemoglobin (HbA) from adult red cells, whereas the more acid-resistant

fetalhemoglobin (HbF) remains intact in fetal red cells. The F cells are stained pink in color

against the normal cells which appear as pale ghost cells and are colorless. It is a reliable

method for identification and quantification of maternal adult cells in the cord blood which is

aspirated in the second trimester of pregnancy for couples undergoing prenatal diagnosis by

fetal blood analysis. The Kleihauer-Betke test (KBT) is widely used for differentiating fetal

erythrocytes from the normal adult cells.

Reagents:

Fixative: 80 % ethanol: 80 mL of absolute ethanol is mixed with 20 mL of distilled

water.

Solution A: Haematoxylin:0.75 g Haematoxylin is dissolved in 100 mL of 96 %

ethanol.

Solution B: Ferric Chloride: 2.4 g Ferric Chloride (FeCl3 anhydrous) is dissolved in

distilled water, 2.0 mL of 25 % HCL is added and the volume is made up to 100 ml.

Page 91: Training Manual for Hemoglobinopathies and Hemophilia:

91

Elution solution :5.0mL of solution A is mixed with 1.0 mL of solution B and

filtered. This solution is prepared freshly and used for elution.

Counter Stain (2.5% Eosin): 2.5 g Eosin is dissolved in 100 mL of distilled water.

Method:

1. A smear is preparedusing a drop of fetal blood on a clean grease free slide and air

dried.

2. It is fixed in 80% ethanol in a coplin jar for 5 minutes.

3. It is then rinsed with tap water and air dried.

4. The slide is placed in the elution solution for 20 seconds; rinsed quickly with water

and counterstained with eosin in another coplin jar for exactly 2 minutes.

5. It is then rinsed with tap water and air dried.

6. The percentage of F- cells is calculated by counting 1000 cells in successive fields

under anoil immersion lens(100X)of the microscope.

Controls:

Mixed control: A drop of fetalblood is mixed with a drop of adult blood, mixed

properly and a smear is prepared.

Negative control: A drop of adult blood with very little HbF.

Positive control: A cord blood sample with a small amount of HbA.

Precaution:

Hematoxylin and FeCl3 can be prepared and stored at room temperature in dark

bottles.

The elution solution should be freshly prepared.

A thin smear is prepared to avoid cell clustering.

Interpretation: Adult cells appear as colourless ghost cells while fetal cells stain pink

A B

Page 92: Training Manual for Hemoglobinopathies and Hemophilia:

92

Figure 32: Fetal cell staining by Kleihauer-Betke method. A) Heterocellular B)

Pancellular

CE-HPLC forsecond trimester prenatal diagnosis of β-Thalassemia and

Sickle Cell Disease by fetal blood analysis

This method can also be used for prenatal diagnosis of β-thalassemia and Sickle cell disease

by fetal blood analysis. Based on the concentration of HbA0, HbF and HbS, one can give the

diagnosis.

Note: The fetal blood obtained by cordocentesis between 18-20 weeks of gestation

should contain 100% of fetal cells.

Preparation of the sample: (EDTA/ Heparinised blood sample)

1 mL of hemolysing solution is mixed with 5 µL of fetal blood and run on the machine.

Interpretation:

Type of hemoglobin Levels of hemoglobin Diagnosis

HbA0 0-<0.5% -thalassemia major

HbA0 >0.5- % -thalassemia trait or normal

HbS < HbA0 HbS trait

HbS >HbA0 Sickle cell disease

Page 93: Training Manual for Hemoglobinopathies and Hemophilia:

93

Figure 33: HPLC chromatograms of fetal blood sample collected during the 2nd

trimester of pregnancy run on the Variant II machine

A distilled water blank is runbefore the fetal blood sample to avoid any carryover.

Thefetal blood samples are always run in duplicate.

Page 94: Training Manual for Hemoglobinopathies and Hemophilia:

94

Extraction of DNA from CVS, AF and Cord blood

Variouskits for DNA extraction from tissues and cells are available commercially (eg.

Qiagen, Invitogen, Genetix etc)

Any of these kits can be used for DNA extraction for CVS, AF and cord blood.

The protocol of DNA extraction should be follow as per the manufacturer’s

instruction

The quality and quantity of the DNA can be checked by methods that are described

earlier in the chapter on Molecular diagnosis of hemoglobinopathies.

Molecular diagnosis of CVS, AF and cord blood

Depending on the mutations of the parents different molecular methods described

earlier such as ARMS, PCR-RFLP and direct DNA sequencing can be used for

detection of mutations in the fetus.

Analysis of Variable Number of Tandem Repeats (VNTRs) using PCR

A variable number tandem repeat (VNTR) is a location in a genome where a short nucleotide

sequence is organized as a tandem repeat. VNTRs are important genetic markers. Due to their

highly polymorphic nature, they are used in gene mapping, paternity analysis, and forensic

medicine and population diversity studies. Herewe describe the use of four different markers

Apo B, D1S80, ACTBP2 and Ig-JH to rule out maternal contamination in chorionic villus

samples for prenatal diagnosis.

Principle:

VNTR analysis is done by using specific pairs of primers complementary to the immediate

flanking region of these genes. It amplifies all the repeats along with the flanking region. The

product obtained by PCR is the function of a number of repeats and it can be resolved by

electrophoresis.

Page 95: Training Manual for Hemoglobinopathies and Hemophilia:

95

Table 19: Different Markers of VNTR

Marker Total no.

of alleles

Repeat

pattern

Allele length

range

Range of no. of

repeats/allele

Apo B 14 30 bp 541-931 bp 27-53

D1S80 16 16 bp 387-723 bp 15-36

ACTBP2 22 4 bp 232-318 bp -

Ig-JH 26 50 520-1720 bp -

Reagents

Table20: Primers used to amplify different VNTR markers

Marker Primer Sequence

Apo B Apo B F (3’) 5’ GAA ACG GAG AAA TTA TGG AGG G 3’

Apo B R (5’) 5’ TCC TGA GAT CAA TAA CCT CG 3’

D1S80 D1S80 F (3’) 5’ GTG TTG TTG GAG ATG CAC GTG CCC CTT GC 3’

D1S80 R (5’) 5’ GAA ACT GGC CTC CAA ACA CTG CCC GCC G 3’

ACTBP2 ACTBP2 F (3’) 5’ ACA TCT CCC CTA CCG CTA TA 3’

ACTBP2 R (5’) 5’ AAT CTG GGC GAC AAG AGT GA 3’

Ig-JH Ig-JH F (3’) 5’ GGG CCC TGT CTC AGC TGG GGA 3’

Ig-JH R (5’) 5’ TGG CCT GGC TGC CCT GAG CAG3’

dNTP Mix

PCR Buffer

MgCl2

DMSO

Taq Polymerase

D/W

Genomic DNA

PCR reactions: Table 21: Reagents used to amplify different markers of VNTR

Reagents Concentration Apo B D1S80 ACTBP2 Ig-JH

Template DNA 100-300 ng 2 µl 2 µl 2 µl 2 µl

Primer 1 (3’) 10 picomole/µl 2 µl 2 µl 2 µl 2 µl

Primer 2 (5’) 10 picomole/µl 2 µl 2 µl 2 µl 2 µl

dNTP mix 25 mM 2 µl 2 µl 2 µl 2 µl

PCR buffer (incomplete) 10X 5 µl 5 µl 5 µl 5 µl

Page 96: Training Manual for Hemoglobinopathies and Hemophilia:

96

MgCl2 25 mM 6 µl 7 µl 9 µl 9 µl

DMSO - - - 5 µl 5 µl

Taq Polymerase 5 Units/ µl 0.5 µl 0.5 µl 0.5 µl 0.5 µl

D/W - 30.5 µl 29.5 µl 22.5 µl 22.5 µl

Total - 50 µl 50 µl 50 µl 50 µl

PCR Programmes:

Table 22: PCR Program for Apolipoprotein B (Apo B)

Denaturation 940C for 5 Mins

25 cycles of

Annealing/Extension 580C for 3 Mins

Denaturation 94 0C for 1 Min

Annealing/ Extension 580C for 5 Mins

Final extension 600C for 10 Mins

Hold 15 0C for

Table 23: PCR program for D1S80(pMCT 118)

Denaturation 950C for 5 Mins

Annealing 650C for 3 Mins

25 Cycles of

Extension 700C for 8 Mins

Denaturation 950C for 1 Min

Annealing 65 0C for 1 Min

Final Extension 700C for 10 Mins

Hold 15 0C for

Table 24: PCR program for ACTBP2(Human - actin related pseudogene H beta

Ac-psi-2)

Denaturation 950C for 5 Mins

Annealing 550C for 2 Mins

30 Cycles of

Extension 720C for 1 Min

Denauration 940C for 20 Secs

Annealing 550C for 30 Secs

Final Extension 72 0C for 5 Mins

Hold 15 0C for

Table 25: PCR program for Ig-JH

Page 97: Training Manual for Hemoglobinopathies and Hemophilia:

97

Denaturation 940C for 5 Mins

Annealing 680C for 3 Mins

25 Cycles of

Denaturation 940C for 2 Mins

Annealing cum Extension 680C for 6 Mins

Hold 15 0C for

Procedure:

1. Add 2 μl of loading dye to the PCRproducts.

2. Load the entire products on a 3% agarose gel.

3. Load the molecular weight marker XIII (Roche) side by side to check specific band

size.

4. Run the gel at 80-100 V for 2-3 hour and view under UV light on a Gel

Documentation System.

Interpretation:

M: Mother; F: Father

Figure 34: 3% agarose gel electrophoresis showingtheD1S80 Marker Family 2: Non-

informative; Family 1,3,4 and 5: Informative (No maternal blood contamination)

References:

1. Kasai K, Nakamura Y, White R, Amplification of Variable number of Tandem Repeat

(VNTR) locus (pMCT118) by the polymerase chain reaction (PCR). And it’s

application to forensic science. J Forensic Sc 1990; 35:11

Centers in different states (Level 3) who will be undertaking prenatal diagnosis will

have to be registered as per the PCPNDT act in their respective states.

Page 98: Training Manual for Hemoglobinopathies and Hemophilia:

98

2. Klehaure E. Determination of fetalhemoglobin: elution technique. In: Schmidt RM,

Huisman THJ, Lehman H (eds). The detection of hemoglobinopathies, CRC Press,

Cleveland, OH. P 20 (1974)

3. Wadia M, Phanasgaonkar S, Nadkarni A, surve R, Gorakshakar A, colah R, Mohanty

D. Usefulness of automated chromatography for rapid fetal blood analysis for second

trimester prenatal diagnosis of beta thalassemia. Prenat. Diagn. 2002;22-153.

Quality Assurance and Equipment Maintenance in Hemoglobinopathies

Laboratory quality can be defined as accuracy, reliability and timeliness of reported test

results. The laboratory results must be as accurate as possible, all aspects of the laboratory

operations must be reliable, and reporting must be timely in order to be useful in a clinical or

public health setting.

Negative consequences of laboratory error

Laboratories produce test results that are widely used in clinical and public health settings,

and health outcomes depend on the accuracy of the testing and reporting. If inaccurate results

are provided, the consequences can be very significant, including:

unnecessary treatment

treatment complications

failure to provide the proper treatment

delay in correct diagnosis

additional and unnecessary diagnostic testing.

These consequences result in increased cost in time and personnel effort, and often in poor

patient outcomes.

Minimizing laboratory error

In order to achieve the highest level of accuracy and reliability, it is essential to perform all

processes and procedures in the laboratory in the best possible way. The laboratory is a

complex system, involving many steps of activity and many people. The complexity of the

system requires that many processes and procedures be performed properly. Therefore, the

quality management system model, which looks at the entire system, is very important for

achieving good laboratory performance.

Page 99: Training Manual for Hemoglobinopathies and Hemophilia:

99

Overview of the quality management system model

1. Organization

In order to have a functioning quality management system, the structure and management of

the laboratory must be organized so that quality policies can be established and implemented.

There must be a strong supporting organizational structure—management commitment is

crucial—and there must be a mechanism for implementation and monitoring.

As the laboratory moves from intent to action in the development of a quality management

system, the major organizational steps will be to assign responsibility for implementation,

allocate resources, develop and distribute a quality manual, begin implementation, and

monitor compliance with the quality policy and the quality management system requirements.

Successful implementation of a quality management system requires planning, management

commitment, understanding the benefits, engaging staff at all levels, setting realistic time

frames and looking for ways to continually improve.

2. Personnel

The most important laboratory resource is competent, motivated staff. The quality

management system addresses many elements of personnel management and oversight, and

reminds us of the importance of encouragement and motivation.

Management of personnel is critical to the success of a quality management programme.

Several elements are important in this management process. Job descriptions should reflect

all skills needed and accurately describe tasks, roles, and authorities. The competency of

personnel will need to be evaluated at the time of hiring and on a regular, recurring basis. A

very important part of the management process is to seek ways to attract qualified personnel,

and to provide motivation and appropriate benefits and working conditions so as to retain

staff.

3. Equipment

Many kinds of equipment are used in the laboratory, and each piece of equipment must be

functioning properly. Choosing the right equipment, installing it correctly, ensuring that new

equipment works properly, and having a system for maintenance are all part of the equipment

management programme in a quality management system.

4. Purchasing and inventory

The management of reagents and supplies in the laboratory is often a challenging task.

However, proper management of purchasing and inventory can produce cost savings in

Page 100: Training Manual for Hemoglobinopathies and Hemophilia:

100

addition to ensuring supplies and reagents are available when needed. The procedures that are

a part of management of purchasing and inventory are designed to ensure that all reagents

and supplies are of good quality, and that they are used and stored in a manner that preserves

integrity and reliability.

A well-managed laboratory will have a system for inventory maintenance and purchasing.

The system will require planning and monitoring to ensure that appropriate quantities of

supplies and reagents are always available, and to prevent wastage.

In implementing an inventory management system, the laboratory must assign responsibility

for the programme, analyze the needs of the laboratory and establish the minimum stock

needed for an appropriate time period. Appropriate logs and forms will be needed, as well as

a procedure for receiving, inspecting and storing supplies. The laboratory will need to

maintain an inventory system for all reagents and supplies used in the laboratory; this system

must include all areas where reagents and supplies are stored.

5. Process control

Process control is comprised of several factors that are important in ensuring the quality of

the laboratory testing processes. These factors include quality control for testing, appropriate

management of the sample, including collection and handling, and method verification

and validation. The elements of process control are very familiar to laboratorians; quality

control was one of the first quality practices to be used in the laboratory and continues to play

a vital role in ensuring accuracy of testing.

a) Sample management components

Written policies for sample management must be established and reflected in the laboratory

handbook. Components to be addressed include:

information needed on requisitions or forms;

handling urgent requests;

collection, labelling, preservation and transport;

safety practices (leaking or broken containers, contaminated forms, other

biohazards);

evaluating, processing and tracking samples;

storage, retention and disposal.

b) Sample processing

Page 101: Training Manual for Hemoglobinopathies and Hemophilia:

101

Verification of quality: Once a sample enters the laboratory, there are a number of steps

needed prior

to testing.

Verify the sample is properly labelled, adequate in quantity, in good condition and

appropriate for the test requested. The test request must be complete and include all

necessary information.

Record sample information into a register or log.

Enforce procedures for handling suboptimum samples, including sample rejection

when necessary.

Rejection of samples: The laboratory should establish rejection criteria and follow them

closely. It is sometimes difficult to reject a sample, but remember that a poor sample will not

allow for accurate results. It is the responsibility of the laboratory to enforce its policies on

sample rejection so that patient care is not compromised.

Management should regularly review the number of rejected samples and reasons for

rejections, conduct training on sample collection, and revise written procedures for sample

management as needed.

The following are examples of samples that should be rejected:

unlabelled sample;

broken or leaking tube/container;

insufficient patient information;

sample label and patient name on the test request form do not match;

haemolysed sample (depending on test requested);

non fasting samples, for tests that require fasting;

sample collected in wrong tube/container (e.g. using the wrong preservative or a

non-sterile container);

inadequate volume for the quantity of preservative;

insufficient quantity for the test requested;

prolonged transport time or other poor handling during transport.

Tracking system: The laboratory needs a system to allow for tracking a sample throughout

the laboratory from the time it is received until results are reported.

This can be done manually by careful keeping of records as follows.

Confirm receipt of samples and include date and time.

Page 102: Training Manual for Hemoglobinopathies and Hemophilia:

102

Label samples appropriately and keep with the test requisition until laboratory

identification is assigned.

Track aliquots—they should be traceable to the original sample.

If computers are available, maintain a database for tracking. The following information about

each sample should be entered into the database:

identification number

patient information

collection date and time

type of sample (e.g. urine, throat, cerebrospinal fluid for culture)

tests to be performed

name of ordering physician (or other health care provider)

location of patient (e.g. ward, clinic, outpatient)

diagnostic test results

time and date results are reported.

c) Sample storage, retention and disposal

Sample storage: Written policies should be developed that include:

description of what samples should be stored;

retention time;

location (consider ease of access);

conditions for storage, such as atmospheric and temperature requirements;

system for storage organization—one method is to store samples by day of receipt

or accession number.

Sample retention: Set a laboratory policy for retention of each type of sample. Some

samples can be quickly discarded and others may need to be retained for longer periods.

Monitor stored samples and do not keep for longer than necessary, as refrigerator and freezer

space may be limited. Sample freeze/thaw cycles must be monitored, as samples may

deteriorate under these conditions.

Sample Disposal: The laboratory is responsible for ensuring that disposal of all laboratory

waste is handled in a safe manner. To ensure proper disposal of patient samples:

Develop a policy for sample disposal; apply local as well as country regulations for

disposal of medical waste.

Establish and follow procedures to disinfect samples prior to disposal.

Page 103: Training Manual for Hemoglobinopathies and Hemophilia:

103

d) Sample transport

Need for transport: Frequently, samples are collected outside the laboratory and must be

transported for subsequent processing and testing. Transport may be for a short distance, but

sometimes a distant clinic or collection site requires the use of vehicles or aeroplanes. In

addition, it may be necessary for the laboratory to ship samples to referral laboratories. In all

cases, transport must be managed carefully in order to maintain integrity of the sample,

giving attention to temperature, preservation needs, special transport containers and time

limitations. It is also important to ensure the safety of those handling the material before,

during and after transport.

Accuracy and precision

If a measurement is repeated many times the result is a mean that is very close to the true

mean.

Accuracy is the closeness of a measurement to its true value.

Precision is the amount of variation in the measurements.

The less variation a set of measurements has, the more precise it is.

In more precise measurements, the width of the curve is smaller because the

measurements are all closer to the mean.

Bias is the difference between the expectation of a test result and an accepted reference

method.

The reliability of a method is judged in terms of accuracy and precision.

A simple way to portray precision and accuracy is to think of a target with a bull’s eye. The

bull’s eye represents the accepted reference value which is the true, unbiased value. If a set of

data is clustered around the bull’s eye, it is accurate.

The closer together the hits are, the more precise they are. If most of the hits are in the bull’s

eye, as in the figure on the left, they are both precise and accurate.

The values in the middle figure are precise but not accurate because they are clustered

together but not atthe bull’s eye. The figure on the right shows a set of hits that are

imprecise.

Page 104: Training Manual for Hemoglobinopathies and Hemophilia:

104

Measurements can be precise but not accurate if the values are close together but do not hit

the bull’s eye. These values are said to be biased. The middle figure demonstrates a set of

precise but biased measurements.

Figure 35: Shows the diagrammatical representation of Accuracy and Precidion.

A QC programme for quantitative tests is essential to ensuring accuracy and reliability of

laboratory testing. The laboratory must establish a QC programme that monitors all

quantitative tests. The programme will have written policies and procedures that are followed

by all laboratory staff.

The overall responsibility of managing the QC programme is usually assigned to the quality

manager, who monitors and reviews all QC data on a regular basis. The recording of the QC

data must be complete and easy to access.

For quantitative testing, statistical analysis can be used for the monitoring process, and the

use of Levey–Jennings charts provides a very useful visual tool for this monitoring.

When controls are out of range, corrective action and troubleshooting must be undertaken;

the problem must be fixed before reporting patient results. Therefore, good protocols for

troubleshooting and corrective action are an important part of the QC process.

Quality control of stains

Some stains can be purchased commercially, but others must be prepared by the laboratory,

following an established procedure. Once stains are made, their bottles should be labelled

with the following information:

name of the stain

concentration

date prepared

date placed in service

expiration date/shelf life

Page 105: Training Manual for Hemoglobinopathies and Hemophilia:

105

preparer’s initials.

Because of their importance, stains should be checked each day of use with positive and

negative QC materials, to make sure their reagents are active and they provide the intended

results. In most cases, positive and negative controls should be stained with each batch of

patients’ slides. All QC results must be recorded each time they are run.

Stains should also be examined to look for precipitation or crystal formation, and to check for

bacterial contamination. Careful maintenance and care of the stock and working solutions of

stains is an essential component in a system to provide good quality in microscopic

examinations.

6. Information management

The product of the laboratory is information, primarily in the form of test reporting.

Information (data) needs to be carefully managed to ensure accuracy and confidentiality, as

well as accessibility to the laboratory staff and to the health care providers. Information may

be managed and conveyed with either paper systems or with computers; both will be

discussed in the section on information management.

Information management is a system that incorporates all the processes needed for effectively

managing data—both incoming and outgoing patient information. The system can be entirely

paper-based, or it can be partly paper-based with some computer support, or it may be

entirely electronic. For either paper-based or computer systems, unique identifiers for patient

samples will be needed. Standardized test request forms, logs and worksheets are also

important to both systems. In helping to prevent transcription errors, a checking process is

beneficial.

When considering adding a computer-based system to a laboratory, cost is a big factor. In

implementation, careful planning and training will help to ensure good results.

A good information management system will:

ensure that all data—the fi nal product of the laboratory—is well managed;

consider all the ways laboratory data will be used when planning a system;

ensure the accessibility, accuracy, timeliness and security of data;

ensure confidentiality and privacy of patient information.

7. Documents and records

Page 106: Training Manual for Hemoglobinopathies and Hemophilia:

106

Many of the 12 quality system essentials overlap. A good example is the close relationship

between "Documents and records" and "Information management". Documents are needed in

the laboratory to inform how to do things, and laboratories always have many documents.

Records must be meticulously maintained so as to be accurate and accessible.

Documents include written policies, processes and procedures, and provide a framework for

the quality system. They need to be updated and maintained.

Records include information captured in the process of performing and reporting a laboratory

test. This information is permanent and does not require updating.

Having a good document control programme ensures that the most current version of a

document is used, and ensures availability and ease of access when a document is needed.

8. Occurrence management

An “occurrence” is an error or an event that should not have happened. A system is needed to

detect these problems or occurrences, to handle them properly, and to learn from mistakes

and take action so that they do not happen again.

Occurrence management is an integral component of laboratory quality management. It

establishes the methods for finding errors and preventing them from occurring again, and also

seeks to identify potential errors and prevent them from happening.

The laboratory should employ an active process for occurrence management and take a

positive approach. Make an effort to detect problems as early as possible, and then take

immediate remedial and corrective action. Be proactive and see opportunities to identify

potential error, thus preventing an occurrence. Finally, keep good records of all problems,

investigations and actions taken.

9. Assessment

The process of assessment is a tool for examining laboratory performance and comparing it to

standards, benchmarks or the performance of other laboratories. Assessment may be internal

(performed within the laboratory using its own staff) or it may be external (conducted by a

group or agency outside the laboratory).

Laboratory quality standards are an important part of the assessment process, serving as

benchmarks for the laboratory.

Page 107: Training Manual for Hemoglobinopathies and Hemophilia:

107

Auditing: The value of a well-designed audit is that it will reveal weaknesses in the pre-

examination, examination and post-examination phases. During audits, information is

gathered about:

processes and operating procedures

staff competence and training

equipment

environment

handling of samples

quality control and verification of results

recording and reporting practices.

The findings are compared with the laboratory’s internal policies and to a standard or external

benchmark. Any breakdown in the system or departure from procedures will be identified.

External quality assessment (EQA): The term EQA is used to describe a method that allows

for comparison of a laboratory’s testing to a source outside the laboratory

Several EQA methods or processes are commonly used. These include:

1. Proficiency testing—external provider sends unknown samples for testing to a set of

laboratories, and the results of all laboratories are analyzed, compared and reported to the

laboratories.

2. Rechecking or retesting—slides that have been read are rechecked by a reference

laboratory; samples that have been analyzed are retested, allowing for inter laboratory

comparison.

3. On-site evaluation—usually done when it is difficult to conduct traditional proficiency

testing or to use the rechecking/retesting method.

Participation in an EQA programme provides valuable data and information, which:

allows comparison of performance and results among different test sites;

provides early warning for systematic problems associated with kits or operations;

provides objective evidence of testing quality;

indicates areas that need improvement;

identifies training needs.

Proficiency testing:

gives a good, objective measure of the laboratory performance

Page 108: Training Manual for Hemoglobinopathies and Hemophilia:

108

can be organized to address most kinds of laboratory testing

is cost-effective and can therefore be used frequently.

Retesting/rechecking:

is useful when it is difficult or impossible to prepare samples to test all of the testing

process;

is expensive and uses considerable staff time.

On-site evaluation:

can give a true picture of a laboratory’s overall performance, and offer real-time guidance

for improvements that are needed;

is probably the most costly, requiring staff time, travel time and expenses of those

performing the evaluation.

10. Process improvement

The primary goal in a quality management system is continuous improvement of the

laboratory processes, and this must be done in a systematic manner. There are a number of

tools that are useful for process improvement.

The process for continual improvement includes:

identification of the problem;

analysis of the data and the processes;

determination of the root cause of the problem;

generation of ideas for solutions.

Continual improvement is the core of quality management, but it requires commitment,

planning, structure, leadership, participation and engagement.

11. Customer service

The concept of customer service has often been overlooked in laboratory practice. However,

it is important to note that the laboratory is a service organization; therefore, it is essential

that clients of the laboratory receive what they need. The laboratory should understand who

the customers are, and should assess their needs and use customer feedback for making

improvements.

Page 109: Training Manual for Hemoglobinopathies and Hemophilia:

109

Seeking customer satisfaction requires commitment from the laboratory management and

staff. It is important to remember that technical competency is not the only goal for the

laboratory. A programme for addressing customer satisfaction requires good planning, the

development of appropriate monitoring tools, and the knowledge to apply the tools to gain

usable information.

Customers or clients of the laboratory include physicians and other health care providers,

hospital and clinic staff, patients and their families, public health officials and the general

community.

Monitoring customer satisfaction requires some resources, primarily involving staff time.

Managers need to ensure that these resources are available.

12. Facilities and safety

Many factors must be a part of the quality management of facilities and safety.

These include:

Security—which is the process of preventing unwanted risks and hazards from entering the

laboratory space.

Containment—which seeks to minimize risks and prevent hazards from leaving the

laboratory space and causing harm to the community.

Safety—which includes policies and procedures to prevent harm to workers, visitors and

the community.

Ergonomics—which addresses facility and equipment adaptation to allow safe and healthy

working conditions at the laboratory site.

When designing a laboratory or organizing workflow, ensure that patients and patient

samples do not have common pathways. To identify where improvements in laboratory

design may be needed in order to prevent or reduce risks of cross contamination, follow the

path of the sample as it moves through the laboratory during the pre-examination,

examination and post-examination phases of testing. The design of laboratory work areas

should ensure proper ventilation and surfaces that can be cleaned and disinfected. In

establishing a safety management programme, it is important to appoint a responsible

supervisor. The laboratory should have a safety manual that establishes policy and describes

standard procedures for handling safety and emergency issues. Personnel need to be trained

in how to apply safety practices and techniques, and to be aware of potential hazards.

Page 110: Training Manual for Hemoglobinopathies and Hemophilia:

110

Quality Assurance for Hemoglobinopathy

1. Hematology Analyzer

For intra‐instrument quality control (internal quality control), the measurement of quality

control samples on a daily basis (or more often) is obligatory. Usually, these quality control

samples are obtained from the manufacturer and they consist of two or three levels (low,

medium, high). There are several pitfalls. First, these samples are usually manipulated to

lengthen the shelf life; therefore, they may behave differently than ordinary patient material.

Second, the manufacturer target's limits are often very broad, and subtle changes in

analyzerbehavior may thus be missed. Therefore, it is recommended to adjust the target range

after a run‐in period of several measurements, for example, to the mean ± 2 standard

deviations. A benefit in using these quality control samples is that they may be used to judge

the instrument precision over time (i.e., drift) using a Levey–Jennings graph; however, it

must be kept in mind that at the end of the shelf life the quality of the control samples may

deteriorate.

An attractive alternative approach is the use of the so‐called moving average. This statistical

method uses the fact that the analytical parameters of the CBC in a large population are stable

over time, changes in the average value thus possibly represent an analytical problem. This

method has proven to be very robust and cheap; however it cannot fully replace the use of

quality control samples.

If a laboratory has multiple HA and/or different analytical techniques that are used for the

same parameter, inter-instrument quality control should be performed. All methods should

lead to the same result in each sample. If different methods lead to unexpected discrepancies

between reported results, this may lead to misinterpretation and possibly unnecessary clinical

intervention. To assure harmonization of reported results between different analyzers and

methods, an inter-instrument quality control comparison is needed. Usually this can be

achieved by measuring patient samples (thereby avoiding so‐called matrix effect of stabilized

quality control samples) on these multiple systems and comparing the results. It is

recommended that multiple HA are compared at least on a weekly basis, using at least three

samples or more.

The participation in an External Quality Assessment (EQA) Scheme (also known as

proficiency testing scheme) is mandatory. The EQA samples should be handled in the same

Page 111: Training Manual for Hemoglobinopathies and Hemophilia:

111

way as routine patient samples to acquire a fair comparison. EQA usually uses manipulated

samples which are composed to resemble pathological samples (e.g., extremely high or low

cell counts). The samples are manipulated to lengthen the shelf life and reduce the sensitivity

of the sample to transport related problems (temperature fluctuations and vibration).

Therefore, direct comparison with patient results is not possible. Laboratories must use the

EQA results to compare their results with (inter)national consensus or reference results and

use these to improve their quality and to harmonize their HA with a consensus group. It must

be kept in mind that, in the absence of a reference method, the results should be compared

with a group of HA that use the same analytical technique, as consensus values may be

systematically biased by inaccuracy of most dominant group method. Moreover, EQA

material should not be used for calibration purposes (because a EQA consensus value and a

true value (calibrator) are not necessarily similar). In some countries, regulatory agencies

abuse the EQA results to evaluate the quality of the participating laboratory. This has a

negative influence on the self‐improvement of the laboratories, and this practice should be

discouraged.

2. High Pressure Liquid Chromatography

Quality assurance Controls should be used with each electrophoretic run, and should contain

the common haemoglobin variants as well as the normal haemoglobins. A control mixture of

HbF, HbA, HbS and HbC is widely used. UK NEQAS (UK National External Quality

Assessment Scheme) circulates samples for the identi¢cation of abnormal haemoglobins and

for the measurement of HbA2, HbF and HbS. Participation in these external quality

assessment programmes is a requirement for Clinical Pathology Accreditation of

haemoglobinopathy laboratories. Commercial controls are available from several

manufacturers for both HbA2 and HbF quantitation and as electrophoretic/chromatographic

markers for haemoglobins such as HbA, HbF, HbA2, HbS, HbC, HbD and HbE. Reference

materials certi¢ed by the World Health Organization are available from the National Institute

of Biological Standards and Controls to assist in the calibration of methods and instruments

used to quantitate HbA2 and HbF

Equipment Management

Page 112: Training Manual for Hemoglobinopathies and Hemophilia:

112

Equipment management is one of the essential elements of a quality management system.

Proper management of the equipment in the laboratory is necessary to ensure accurate,

reliable and timely testing.

The benefits of a good equipment management programme are many:

helps to maintain a high level of laboratory performance;

reduces variation in test results, and improves the technologist’s confidence in the

accuracy of testing results;

lowers repair costs, as fewer repairs will be needed for a well-maintained

instrument; lengthens instrument life;

reduces interruption of services due to breakdowns and failures;

increases safety for workers;

produces greater customer satisfaction.

A great deal of thought and planning should go into equipment management. As the

laboratory puts an equipment management programme in place, the following elements

should be considered.

Selection and purchasing—when obtaining new equipment, what criteria should be used to

select equipment? Should equipment be purchased or would it be better to lease?

Installation—For new equipment, what are the installation requirements and who will

install the new instrument?

Calibration and performance evaluation—What is needed to calibrate the equipment and

validate that it is operating correctly? How will these important procedures be conducted for

both old and new instruments?

Maintenance—What maintenance schedule is recommended by the manufacturer? Will

the laboratory need additional preventive maintenance procedures? Are current maintenance

procedures being conducted properly?

Troubleshooting—Is there a clear procedure for troubleshooting for each instrument?

Service and repair—What is the cost? Can the laboratory obtain the necessary service and

repair in its geographical area?

Retiring and disposing of equipment—What must be done to dispose of old equipment

when it needs to be replaced?

It is the responsibility of the laboratory director to:

oversee all the equipment management systems in the laboratory;

Page 113: Training Manual for Hemoglobinopathies and Hemophilia:

113

ensure that all persons who will be using the instruments have been appropriately trained

and understand how to both properly operate the instrument and perform all necessary routine

maintenance procedures.

Equipment management responsibility may be specifically assigned to a technologist in the

laboratory. In many laboratories, there is a person who has good skills with equipment

maintenance and troubleshooting. Giving this person the role of oversight of all equipment is

recommended.

Oversight of an equipment management programme includes:

assigning responsibilities for all activities

ensuring that all personnel are trained in operation and maintenance

monitoring the equipment management activities, including

- reviewing all equipment records routinely

- updating maintenance procedures as necessary

- ensuring that all procedures are followed.

Equipment Maintenance

1. Preventive maintenance

Preventive maintenance includes measures such as systematic and routine cleaning,

adjustment and replacement of equipment parts at scheduled intervals. Manufacturers

generally recommend a set of equipment maintenance tasks that should be performed at

regular intervals: daily, weekly, monthly or yearly.

Following these recommendations will ensure that the equipment performs at maximum

efficiency and will increase the lifespan of the equipment. This will also help to prevent:

inaccurate test results due to equipment failure

delays in reporting results

low productivity

large repair costs.

2. Equipment Calibration

Follow the manufacturer’s directions carefully when performing the initial calibration of the

instrument. It is a good idea to calibrate the instrument with each test run, when first putting

it into service. Determine how often the instrument will need to be recalibrated, based on its

stability and the manufacturer’s recommendation. It may be advantageous to use calibrators

provided by or purchased from the manufacturer.

Page 114: Training Manual for Hemoglobinopathies and Hemophilia:

114

3. Performance evaluation

Prior to testing patient specimens, it is important to evaluate the performance of new

equipment to ensure it is working correctly with respect to accuracy and precision. In

addition, test methods using kits or laboratory instruments need to be evaluated for the ability

to detect disease (sensitivity, specificity, positive and negative predictive value) and to

determine normal and reportable ranges.

Verification of manufacturers’ performance claims—Manufacturers provide performance

evaluations for testing methods using their kits or instruments, and include the information in

the package inserts or operator's manuals. However, laboratories need to verify the

manufacturer's performance claims, and demonstrate they can get the same results using the

kits or equipment in their laboratory, with their personnel.

Some of the steps that should be followed to verify performance include:

testing samples with known values and comparing the results to the expected or certified

value;

if equipment is temperature controlled, establishing the stability and uniformity of the

temperature.

Validation of new equipment and associated techniques—If the equipment and associated

techniques are new, validation processes will be important. Validation can be carried out by

running samples in parallel using both old and new equipment and methods for a period of

time to determine that the expected results can be obtained. These validation procedures

should be completely recorded.

4. Maintenance plan

A maintenance plan will include preventive maintenance procedures as well as provision for

inventory, troubleshooting and repair of equipment. When implementing an equipment

maintenance program, some of the initial steps will include:

assigning responsibility for providing oversight;

developing written policies and procedures for maintaining equipment, including routine

maintenance plans for each piece of equipment that specify the frequency with which all

maintenance tasks should be performed;

developing the format for records, creating logs and forms, and establishing the processes

to maintain records;

Page 115: Training Manual for Hemoglobinopathies and Hemophilia:

115

training staff on the use and maintenance of the equipment, and ensuring that all staff

understands their specific responsibilities.

It is recommended that a label is attached to the instrument indicating when the next

maintenance or service should be performed. The laboratory should keep an inventory log of

all equipment in the laboratory. The log should be updated with information on new

equipment and include documentation of when old equipment is retired. For each piece of

equipment, the equipment inventory log should have a record of:

instrument type, make and model number, and serial number so that any problems can be

discussed with the manufacturer;

date the equipment was purchased, and whether it was purchased new, used or

reconditioned;

manufacturer/vendor contact information;

presence or absence of documentation, spare parts and maintenance contract;

warranty’s expiration date;

specific inventory number indicating the year of acquisition (this is especially useful for

larger laboratories); for example, use the style “YY-number” (04-001, 04-002, etc.) where

“YY-number” equals the last two numbers of the year followed by a number attributed in the

year.

An inventory process must be conducted if the laboratory does not have an existing inventory

system for equipment. This could be conveniently organized following a model grid, room by

room; for example, conduct an inventory of equipment in the reception area, then the sample

collection area, the serology testing area, and the parasitology testing area. During the

inventory, the condition of the equipment should be documented as functional, partially

functional or nonfunctional. Equipment that is not functioning needs to be evaluated as

towhether or not it can be repaired. Nonrepairable equipment should be retired, and work

should be scheduled for equipment needing repair.

5. Inventory of spare parts

To ensure that the laboratory does not run out of spare parts, an inventory record of those

used most frequently should be kept for each piece of equipment.

The record should include:

part name and number;

average use of the part, and the minimum to keep on hand;

Page 116: Training Manual for Hemoglobinopathies and Hemophilia:

116

cost;

date when the part is placed into storage and when it is used (in and out stock log);

quantity of each part remaining in inventory.

6. Maintenance documentation

Equipment documents and records are an essential part of the quality system. The policies

and procedures for maintenance should be defined in appropriate documents, and keeping

good equipment records will allow for thorough evaluation of any problems that arise

Each major piece of equipment will have its own equipment maintenance document. Smaller,

commonly used equipment such as centrifuges and pipettes may be managed with an

equipment maintenance document or manual that deals with all such equipment in the

laboratory. An equipment maintenance document should include:

step-by-step instructions for routine maintenance, including frequency of performance and

how to keep records of maintenance;

instructions for carrying out function checks, frequency of performance, and how to record

the results;

directions for calibrating the instrument;

guide for troubleshooting;

any required manufacturer’s service and repair;

list of any specific items needed for use and maintenance, such as spare parts.

For major equipment, include identification of the specific instrument and perhaps

information on its performance.

Each piece of equipment should have a dedicated logbook documenting all characteristics

and maintenance elements, including:

preventive maintenance activities and schedule;

recording of function checks and calibration;

any maintenance performed by the manufacturer;

full information on any problem that the instrument develops, the subsequent

troubleshooting activity and follow-up information regarding resolution of the problem. In

recording problems, be sure to record

- date problem occurred and when equipment was removed from service;

- reason for breakdown or failure;

- corrective action taken, including a note about any service provided by the manufacturer;

Page 117: Training Manual for Hemoglobinopathies and Hemophilia:

117

- date returned to use;

- any changes to procedure for maintenance or function checks as a result of the problem.

Hematology Analyzer maintenance

Validation: According to ISO 15189, the laboratory should only implement HA that are

validated for the intended purpose.

First, certain parameters must be compared to a reference method. Most of the current

reference methods are outdated such as hematocrit, red blood cell count, white blood cell

count, and white blood cell differential. In such cases, improved performance of the new

method is compared or calibrated to a relatively poor standard, which leads to a poorer

performance than analytical and technically feasible. Unfortunately, in some countries, the

registration authorities demand a comparison with these reference methods and/or with a

current generation analyzer. This requirement could thus lead to poorer analytical

performance than necessary and also hampers innovation.

Second, it is challenging to compare the ‘flagging’ of a new instrument with an existing

method. Samples are for instance ‘flagged’ to identify pathological samples that need rerun in

a different mode, revision, or additional diagnostic work‐up (i.e., microscopic review or flow

cytometry). The underlying algorithms that generate these flags may be totally different,

making a comparison between two types of HA challenging. Usually manufacturers try to

make sure that all pathological samples are recognized, for example, the samples with

malignant WBCs (blasts). To avoid missing these pathological samples, the flagging

algorithm generates flags with a low positive predictive value (PPV) [and usually a high

negative predictive value (NPV)], that is, a lot of normal samples get a suspicious flagging

and need (microscopic) review, thereby unnecessary increasing the workload of the

laboratory.

Lastly, establishing reference intervals by manufacturers can be arduously, especially in the

pediatric population. Obtaining samples from healthy children and neonates, has shown to be

difficult. Reference intervals in the pediatric population are often based on publications from

decades ago, using technology that is no longer widely available, making it questionable

whether these reference ranges can be applied in the present time; or there are new

parameters or parameters using a new analytical technique for which there are no published

Page 118: Training Manual for Hemoglobinopathies and Hemophilia:

118

reference ranges available. Therefore, manufacturers should make considerable efforts to

generate reference intervals in these groups for their new HA.

Verification

The ISO 15189 states that the laboratory shall verify upon installation and before use that the

equipment is capable of achieving the necessary performance and that it complies with

requirements relevant to any examinations concerned. Both the ICSH and CSLI have

developed guidelines for the verification of HA. According to these guidelines, verification

studies should be performed on precision (reproducibility), accuracy (method comparison),

analytical sensitivity (limit of detection), analytical specificity including interfering

substances, reference ranges, patient correlation studies, linearity, limits of detection,

carryover, and the analytical measuring range. However, the ICSH and CLSI guidelines do

not specify the limits of acceptability of these studies throughout the different parameters.

Nor do these guidelines suggest a minimal range in which the reliability of the parameters

must be established. The extensiveness of the verification study depends upon the availability

of independent evaluation data (preferably published in the peer‐reviewed literature), the

extent of CBC parameters reported by the laboratory, and the range of samples available for

the evaluation of the equipment. As hematology parameters include cell differentiations and

cell indices, some (pre)analytical characteristics differ from more regular laboratory tests

expressed per concentration plasma. Therefore, some in‐depth knowledge about hematology

and hematology parameters is required to interpret the verification analyses.

Before starting a verification process, we should establish the medically allowable error. The

combination of different aspects of test performance forms the total analytical error. The total

analytical error reflects the analytic reliability of a test result. If a laboratory wants to verify

that it produces reliable results in a clinical perspective, there are several considerations.

Firstly, it is advised to establish which levels are clinically relevant. For instance, if platelet

transfusions are given below a platelet count of 10 × 109/L, it is sensible to verify the test

performance around this concentration. Secondly, the total analytical error around this cut off

point should be acceptable for clinical decision making. This medically allowable error may

depend on the expectations of the clinicians, the biological variation, and the current

state‐of‐the‐art performance.

Page 119: Training Manual for Hemoglobinopathies and Hemophilia:

119

Precision

Precision is the closeness of agreement between repeated measurements of a sample.

Within‐run precision (also known as reproducibility or repeatability) usually consists of a

single run of 20 measurements and is reported as a coefficient of variation [CV (%)].

Between‐batch precision [reported as CV (%)] is based on single measurements that are

repeated every day for a 20‐day period and is affected by random error and drift. Usually,

stabilized quality control samples are used to establish the between‐batch precision. The

manufacturers' claims about both within‐run and between‐batch precision must be verified. In

addition, we should always attempt to reach at least the current state‐of‐the‐art CV (%) for

reproducibility, so we can provide physicians with the most accurate information and thereby

provide opportunities to improve medical decision making and quality of care.

Accuracy and comparability

Accuracy (also known as trueness) is used to describe the closeness of a measurement to the

true value. The use of a ‘true value’ for the CBC is hard to apply in daily laboratory practice,

as reference methods only exists for a few selected parameters and these reference methods

tend to be rather impractical. For the leukocyte differential count, the microscopic evaluation

of a slide is currently regarded as the reference method (400‐cell manual differential).

However, this method suffers from several disadvantages such as statistical error, slide

distribution error, and morphological interpretation error. As a surrogate, usually in a

verification process, the HA under evaluation is compared with the instrument in routine. In

the comparability study (patient correlation studies), as many samples as possible (generally

400 samples or more for each parameter) should be compared using both HA. Furthermore, it

is of importance to include normal and abnormal samples in approximate equal proportions.

The difference between the HA under evaluation and the current analyzer or reference

method should be as low as possible. The parameter from the HA under evaluation can be

compared with the current HA using linear regression. Perfect concordance results in a

correlation coefficient (r value) of 1. Yet, if the new HA is much more accurate than the old

HA, this will negatively affect the correlation coefficient, and thus, the correlation coefficient

should be interpreted with caution. In addition, some parameters are known for their poor

correlation, such as the basophil count.

HPLC Maintenance

Page 120: Training Manual for Hemoglobinopathies and Hemophilia:

120

HPLC is the sensitive and high cost equipment. So, we need to care about regular

maintenance of HPLC. It leads to reducing the cost due to routine problems. The equipment

should be inspected weekly for signs of leaks. Prior to any analysis, a system suitability test,

which closely resembles the intended assay, should be performed to ensure that the system is

operating within establish criteria. Before using mobile phase solvents one should be

thoroughly familiar with hazards and safe handling practices. Observe the manufacturer

recommendations for use, storage and disposal. These recommendations are normally

provided in material safety data sheets supplied with the solvents. The storage condition is

one of the important parameter to maintain the HPLC. One should not operate the system in a

cold room or refrigerated area. The ambient temperature is 10-35◦C, ambient relative

humidity is 20-80%. It may vary depending on manufacturers. Some ofthe basic assumptions

to maintain HPLC are, the HPLC is plugged in and turned on, solvent is in the reservoir, The

detector hasa good lamp in it, the solvent bottle does not have a vacuum on it, should not use

acetone at 195nnm, should not use the water and hexane as gradient, should not use methanol

and water without degassing, solvent pH should not exceed 13 on a silica base column,

should not blow HCL vapours into HPLC, should not flush system with methanol after

running buffer, do not filter organic solvents through organic solvents, do not try to change

the column frits while it still has pressure in it, do not pump cyclohexane above 2000 psi and

don’ttighten the mobile phase container. The instrument should be frequently

calibratedusingtheappropriate procedure.

Calibrators and Controls

All HPLC runs are preceded by priming and then calibration of theinstrument. Separate

calibration factors are obtained for HbA2 and HbF as ratios of expected to obtained values.

Since the two values should ideally be equal (i.e., a ratio of 1) these are deemed to have

passed if they lie between 0.7 and 1.3. These calibration factors are then applied for all

subsequent patient samples. The retention time of HbA2 in the calibrator is also a useful

indicator of run reliability. Normally it lies between 2.60-2.70 min, and the instrument may

need temperature adjustments if wider deviations occur. This is especially common as the

column cartridge ages; usual cartridge lifetimes being around 250 injections. Bi-level

controls, one normal (HbF 1%-2%, HbA2 1.8%-3.2%) and one elevated (HbF 5%-10%,

HbA2 4%-6%), should be analyzed at the beginning as well as the end of each set of patient

specimens. The high control in case of Bio-Rad instruments also contains a variant peak that

Page 121: Training Manual for Hemoglobinopathies and Hemophilia:

121

must elute in the S-window. All peaks must be symmetrical, temperature variations being the

most common cause again of asymmetry.

References

1. ISO 9000:2005. Quality management systems–fundamentals and vocabulary. Geneva,

International Organization for Standardization, 2005.

2. CLSI. User protocol for evaluation of qualitative test performance, approved

guideline—2nd ed.

3. Cochran C. The five keys to a successful internal audit program. The Auditor 2:1.

Chico, CA, Paton Press, 2007 (http://www.dnvcert.com/DNV/Certifi

cation1/Resources1/Articles/NewsletterInfo/FiveKeystoaSuccessfulI/).

4. Kusum M, Silva P. Quality standards in health laboratories implementation in

Thailand: a novel approach. World Health Organization Regional Office for South-

East Asia, 2005 (http://www.searo.who.int/LinkFiles/Publications_SEA-HLM-

386__a4___2_.pdf

5. WHO.Accreditation of health laboratories in the countries of the SEA region: report

of a regional consultation, Bangkok, Thailand, 6–10 October, 2003. WHO Project:

ICP BCT 001, World Health Organization Regional Office for South-East Asia, 2004.

6. WHO Quality Assurance in Hematology 1998

7. Statistical methods for proficiency Testing ( NABL& Indian Statistical Institute)2003

8. Interlaboratory tests in :- Wenclawiak, Koch, Hadjicostas (eds). Quality Assurance in

Analytical Chemistry – Training and Teaching (2nd Edition) Springer verlag. Berlin

Heidelberg 2010

9. ISO 13528:2015, Statistical methods for use in proficiency testing by inter-laboratory

comparison: International standard.

10. Laboratory quality management system: handbook of WHO2011

Page 122: Training Manual for Hemoglobinopathies and Hemophilia:

122

Genetic Counseling for Hemoglobinopathies

Genetic counseling can be defined as:

A communication process by skilful counselors or medical social workers who have had

adequate training to provide often difficult to comprehend information and support to

individuals and families about genetic risks, testing and diagnosis of hemoglobinopathies,

consequences of the disorder and ways of preventing further birth of affected children with an

inherited hemoglobin disorder in the family.

Genetic counseling should help families understand and adapt to the medical,

psychological, and familial implications of the disease, or carrier status.

A trained genetic counselor can provide many important functions for the family;

however we have few such experts in the country. Armed with important and complete

information about the disorder, other trained healthcare professionals can also provide

this valuable support.

Genetic counseling is required:

Before undergoing screening for a hemoglobinopathy trait.

After diagnosis of a carrier state or major disease at different stages.

Prior to pregnancy and/or as early in pregnancy as possible.

After prenatal diagnosis to inform the couple about the diagnosis of the fetus.

Prior to and after newborn screening for a hemoglobinopathy eg sickle cell disease.

Pre and post hematopoietic stem cell transplant.

As an ongoing process rather than a onetime communication.

Awareness is the key to a successful programme:

Inspite of efforts since several years by many groups, awareness on the thalassemias and

sickle cell disorders is very limited in the population. Awareness generation is needed both

among medical professionals particularly pediatricians, obstetricians and family physicians,

among nurses, health care workers as well as different sections of society including school

and university students, young adults of marriageable age, newly married couples before

conception and during early pregnancy. With a better understanding of the inherited

Page 123: Training Manual for Hemoglobinopathies and Hemophilia:

123

hemoglobin disorders, their inheritance and consequences, screening and genetic counseling

will have greater meaning. An accurate diagnosis is a prerequisite for counseling.

Questions couples with an affected child have:

Why is our child suffering from the disease when we are both normal?

What treatment options are available and is there a cure for the disease?

What is the cost involved for treatment?

Can the next child also have a similar disease?

Is there any way to prevent it?

Components of genetic counseling:

Helping to proactively prepare the couple for the possibility of having a child with an

inherited hemoglobinopathy.

Interpretation of family and medical histories to assess the chance of disease

occurrence or recurrence.

Explaining the clinical presentation, severity and consequences of the disorder.

Explaining the autosomal recessive inheritance pattern.

Explaining to the family the risks and meaning of the carrier state.

Drawing a pedigree chart over three-generations to record family history, noting

consanguinity and ethnicity.

Promoting informed choices and adaptation to the risk or condition.

Explaining other options- providing information about natural, assisted, and non-

reproductive opportunities available for family planning.

Assessing risk for thalassemia, sickle cell disease or other compound heterozygous

conditions in family members.

Providing emotional and social support to reduce the family's anxiety.

Helping patients and parents to convey information about the genetic risk to other

family members.

Taking an informed consent.

Both pre and post-test counseling are important to enable the individual or family to

face the situation and take an appropriate decision.

Page 124: Training Manual for Hemoglobinopathies and Hemophilia:

124

During pre-test counseling, the clinical presentation of the condition the patient may be at

risk for is discussed, the pattern of inheritance of the condition, risk of recurrence, available

testing procedures with their advantages and limitations, reproductive options available and

the required follow up.

The inheritance patterns:

This should be explained by the counselor with the help of pedigree charts showing different

matings as shown below (Figures 36-40). The same inheritance pattern holds true for both the

thalassemias and sickle cell disease as well as other genetic combinations where one parent

is a carrier of thalassemia and the other is a carrier of a variant hemoglobin like HbS, HbE,

HbD or the uncommon HbLepore.

Figure 36 - One parent is normal and the other is a thalassemia carrier or a sickle cell

carrier. There is a 50% chance in every pregnancy that the baby will be normal and a

50% chance that the baby will be a thalassemia carrier or a sickle cell carrier.

Page 125: Training Manual for Hemoglobinopathies and Hemophilia:

125

Figure 37 -Both parents are thalassemia or sickle cell carriers. There is a 25% chance

in every pregnancy that the baby will be normal, a 50% chance that the baby will be a

carrier like the parents and a 25% chance that the baby will be affected with

homozygous thalassemia or sickle cell anemia.

Figure 38 - One parent is homozygous for β-thalassemia or has sickle cell anemia and

the other is normal. In every pregnancy the baby will be a β- thalassemia carrier or a

sickle cell carrier.

Page 126: Training Manual for Hemoglobinopathies and Hemophilia:

126

Figure 39 - One parent is homozygous for thalassemia or has sickle cell anemia and

the other is a thalassemia carrier or sickle cell carrier. There is a 50% chance in every

pregnancy that the baby will be a thalassemia carrier or a sickle cell carrier and a

50% chance that the baby will have homozygous thalassemia or sickle cell anemia.

Figure 40 -The very rare possibility that both parents have homozygous thalassemia

or sickle cell anemia. In such a situation in every pregnancy the baby will have

homozygous thalassemia or sickle cell anemia.

Due to the considerable phenotypic diversity of the thalassemia syndromes, it is imperative

that the genetic counselor has adequate knowledge of the molecular mechanisms involved in

Page 127: Training Manual for Hemoglobinopathies and Hemophilia:

127

this diversity of clinical presentation ranging from a very severe thalassemia major disorder

to a mild thalassemia intermedia presentation. The counselor should also be aware of other

ameliorating factors like the co-inheritance of thalassemia or gene triplication and

variations in the gene in modulating the disease apart from the inheritance of severe or mild

thalassemia mutations. These factors must also be discussed with the families seeking

advice.

Counseling couples at risk for sickle cell disease is also quite complex because of the variable

severity of sickle cell disorders, which can also vary from an early presentation with repeated

complications in some, to a relatively mild or even an almost asymptomatic presentation in

others. This must be clearly explained to the parents although it makes it considerably

difficult for them to take a decision on whether or not to opt for prenatal diagnosis.

Variable disease severity:

The table below (Table 26) shows the severity of homozygous or compound heterozygous

babies when different combinations of genes are inherited.

Table 26: The severity of homozygous or compound heterozygous babies

Homozygous thalassemia Usually severe thalassemia major; in 10 to 15% milder

thalassemia intermedia

Sickle cell anemia Variable clinical severity; more severe presentation in some

population groups than in others

Homozygous Hb E Mild clinical presentation

Homozygous HbD Very mild to asymptomatic presentation

HbS - thalassemia Usually a severe clinical presentation

HbE - thalassemia Often a severe clinical presentation

HbD - thalassemia Generally a mild presentation

Hb Q - thalassemia Very mild to asymptomatic presentation

thalassemia - thalassemia Moderate to severe clinical presentation

HbSD disease Severe clinical presentation

HbSE disease Usually a severe clinical presentation

HbDE disease Usually a milder clinical presentation

thalassemia – HPFH Mild clinical presentation

Page 128: Training Manual for Hemoglobinopathies and Hemophilia:

128

The general principles of counseling must be followed:

Ensure a quiet room where the parents of the child can sit and ask questions without

interruption.

Prepare the family by explaining that you have some important information regarding

the child. Include other family members if parents wish to have them present.

Explain in a language that the family can understand, avoid technical terms. If using

scientific terms or when explaining disease explain each term, repeat, ask if

understood before going on.

Pause and ask if any questions, frequently.

At the end of counseling, give written information sheet tothe family and ask them to

return it if any other questions.

Parents and families who are given a diagnosis of a genetic condition may have anger

and resentment. They often go through a period where they may be in denial.

Sometimes the anger may be targeted at the person giving them the information.

Hence information about diagnosis should be provided by the doctor /and an

additional staff.

Ethical and Religious Issues:

Apart from the information given by the genetic counselor, the acceptability of prenatal

diagnosis with selective termination of an affected fetus by couples is also guided by social,

cultural and religious factors as well as the influence of elders in the family. Genetic

counselling should be “non-directive”, where the genetic counselorshould present all the facts

and information and after understanding and considering all options, the couple or family

take a decision without being influenced by the counselor.

Ethical practice requires the following:

Protecting the right to autonomy of the individual or couple to make an informed

decision.

Their right to get complete information from the counselor.

Confidentiality should be maintained and their privacy protected to avoid

stigmatization.

Page 129: Training Manual for Hemoglobinopathies and Hemophilia:

129

Life-expectancy for individual’s withhalassaemia has improved significantly with

improved management protocols, and in most countries Quality of Life (QoL) is now

considered an important factor of effective health care. These issues on the available

treatment and QoL should also be discussed during genetic counseling.

Informed Consent:

Informed decision making is most often suboptimal particularly when the literacy level of the

individuals or family is low as is often the case in remote rural regions of the country. The

counselor has to bear this in mind while explaining the need for genetic testing and the

consequences of having a child with an inherited hemoglobin disorder.

Newborn screening for hemoglobinopathies allows presymptomatic diagnosis and timely

management of sickle cell disease. Counselors must explain the purpose of newborn

screening to the parents before taking an informed consent from them to test their newborn

baby. At the same time it provides an opportunity for genetic counseling of parents which

would help them to take reproductive decisions in subsequent pregnancies.

For couples opting for prenatal diagnosis, it is crucial to investigate and assess the couple

before pregnancy whenever possible. This will permit the laboratory to undertake molecular

analysis and know the mutations in the parents before the intervention is undertaken. It is also

important to explain and make the couple aware of the risks and complications of the fetal

sampling procedure and also about the occasional problems of the need of repeating the

procedure, if for technical reasons the laboratory is unable to give a report and also the very

occasional problem of a misdiagnosis.

The genetic counselor has to take an informed consent before any genetic screening or

testing is done or any intervention procedure is undertaken. Consent can be written, verbal or

non-verbal/implied as is followed in the UK and the written consent form serves as evidence

that consent has been given. Before this, the benefits, risks and other available options must

be explained to the patient. There are often privacy issues in a hospital environment and

counseling must be given to individual families in a confidential manner.

References

1. Colah R, Italia Y, Gorakshakar A. Burden of thalassemia in India: The road map for

control. PediatrHematol Oncol J. 2018; DOI: 10.1016/j.phoj.2017.10.002

Page 130: Training Manual for Hemoglobinopathies and Hemophilia:

130

2. Colah R, Italia K. Genetic Counseling for Hematological Disorders. In Fetal and

Neonatal Hematology, Oncology and Immunology Eds Lokeshwar MR, Suchdeva A,

Shah NK, Agarwal B, Manglani M. Jaypee Brothers Medical Publishers 2017; 28-42

3. Mohanty D, Das K. Genetic counselling in tribals in India. Indian J Med Res.

2011;134:561-71.

4. Puri RD, Verma IC. Screening, genetic counselling, prenatal diagnosis and pre-

implantation diagnosis for thalassemia. In: Sachdeva A (Ed). Thalassemia: National

guidelines for management of transfusion dependent and non-transfusion dependent

thalassemia, New Delhi. 2014. pp. 272-84.

5. Baker, DL, Schuette JL, Uhlmann WR. A Guide to Genetic Counseling. New York:

Wiley-Liss, 1998

6. Colah RB, Mukherjee MB, Ghosh K. Sickle cell disease in India. Current

OpinHematol. 2014;21:215-23.

7. Muthuswamy V. Ethical issues in genetic counselling with special reference to

haemoglobinopathies. Indian J Med Res. 2011;134:547-51.

8. Deka D, Malhotra N. Genetic counselling for obstetricians. Donald School J

Ultrasound Obstet Gynecol. 2010;4:447-53.

9. Bhat M. Social and cultural issues in genetic counselling. J Biosci. 2015;40:217-20.

10. Giordano PC. The Genetic Counselor. In: Carrier diagnostics and prevention of

hemoglobinopathies using high-performance liquid chromatography. Companion

handbood for the physician, the laboratory doctor and the genetic counselor. Hercules,

CA: Bio-Rad Laboratories Inc; 2006. pp. 81-93.

Page 131: Training Manual for Hemoglobinopathies and Hemophilia:

131

SECTION 2

MANAGEMENT OF HEMOGLOBINOPATHIES

Page 132: Training Manual for Hemoglobinopathies and Hemophilia:

132

2. Management of Hemoglobinopathies

i. Thalassemia Major

Basic recognition of symptoms and signs of thalassemias at level 1

(PHC/CHC)

Thalassemia is an inherited disease of the blood, passed from parents to children through the

genes. This hereditary disorder occurs as a result of mutations affecting the synthesis of the

hemoglobin molecule, found in the red blood cells (RBCs).

In ‘normal’ or adult hemoglobin (HbA), a protein is formed consisting of two alpha (α) and

two beta (β) chains. In thalassemia, there is a reduction in the production of either α- or β-

chains in the hemoglobin molecule.

In α-thalassemia, there is a decrease or lack of

α-chains.

The genes responsible for the production of α-

chains are the α-globin genes, located on

chromosome 16

In β-thalassemia, there is a decrease or lack of

β-chains.

The genes responsible for the production of β-

chains are the β-globin genes, found on

chromosome 11

Classification of Thalassemia disease states is based on severity: The diagnosis of

Thalassemia state has a wide range of clinical severity, however they are broadly divided into

two broad categories.

Page 133: Training Manual for Hemoglobinopathies and Hemophilia:

133

A. Thalassemia Major (Transfusion Dependant Thalassemia): Patients in this category

cannot survive for long after birth without regular blood transfusions and require

regular transfusions beginning at 6 to 24 months of age for sustaining their lives. A

later diagnosis ( >3 years)should make us suspect thalassemia intermedia. This is

important as some patients labeled as thalassmia major may respond to Hydroxyurea

if they are thalassemia intermedia

B. Thalassemia Intermedia[Non Transfusion Dependant Thalassemia (NTDT)]: Patients

in the second category, thalassemia intermedia, may survive with occasional or no

transfusions, or with chronic transfusions initiated at an older age. A patient initially

labeled as NTDT may later require transfusions and even regular transfusion therapy

if they have persistent growth retardation in consultation with a hemoglobinopathy

specialist.

Differences in the severity of thalassemia are usually due to genetic factors, such as

mutations that result in the production of more β-globin chains, or the co-inheritance of α-

thalassemia and/or other genetic factors, each of which contribute different mechanisms in

ameliorating the pathology and clinical outcome of the disease. For example, a given factor

may allow for the production of some fetalhemoglobin (HbF), leading to the partial

correction of the imbalance occurring between the α- and β-chains, which is the single most

important cause of the pathophysiology of thalassemia.

Table 1.

When to Suspect Thalassemia?

History :

Presentation :6 to 24 months age

Lethargy

Progressive Pallor

Failure to thrive

Feeding problems, abdominal distension

Irritability

Recurrent Fever

Family history of low Hemoglobin

sibling death or blood transfusionhistor

Examination :

History :

Presentation : 2 to 6 years of age or later

Mild Pallor

Mild Jaundice

Family history of low hemoglobin

Or sibling death

Examination :

Page 134: Training Manual for Hemoglobinopathies and Hemophilia:

134

Pallor,

Growth retardation, pallor,

Jaundice,

Palpable liver and spleen , leg ulcers,

Skeletal and dental deformities

Pallor,

Hepatosplenomegaly,

Skeletal and dental deformities

Extramedullary masses

Suspect

Thalassemia major also known as

Transfusion Dependent Thalassemia (TDT)

Suspect

Thalassemia intermedia now called –

Non Transfusion Dependent

Thalassemia (NTDT)

Get Complete Blood Count in an automated cell counter with RBC indices

Peripheral Blood Smear

Initial Stabilization (see next section)

Refer to level 2 if tests Hb<7g/dl

( MCV 50-70fl; MCH: 12-20pg suggestive of Thalassemia)

Do’s and Don’ts for management and referral for a suspected case of Thalassemia

Do’s Don’t’s

Do a thorough clinical examination

and detailed history

Do not transfuse if diagnosis is not

confirmed and the patient is clinically

stable.

Document the history, family and

examination findings

If transfusion is deemed urgent

preserve a sample for baseline

diagnostic testing (HPLC) (Refer to

section on transfusion for details)

Give initial stabilization and basic

medical investigation

Do not delay referral if patient is sick

Counsel the family about possible

diagnosis, need for investigations and

future course

Send the blood sample for blood

grouping, cross matching

The first diagnosis and long term

management plan should be

ascertained in consultation with

hemoglobinopathy expert

Page 135: Training Manual for Hemoglobinopathies and Hemophilia:

135

Initial work up, stabilization and triage of care of the sick

patient(see diagnostic section toofor tests to be done)

Age of presentation:

Thalassemia major patients usually are diagnosed between 6 months to 2 years of age.Though

rarely symptoms may start as early as 3 months or even occur after two years of age.

Newborns are asymptomatic because of HbF, which is produced by gamma globin chains, at

the time of the normal switch in hemoglobins from fetal (HbF) to adult hemoglobin (HbA),

the defect manifests as anemia due to defect in beta globin synthesis required for production

of normal adult hemoglobin. If untreated the mortality of thalassemia major is very high, it is

estimated that 85 percent of untreated children will die before the age of five years.

Thalassemia intermedia patients usually present later in life due to milder phenotype than

major, the clinical features start to manifest over 2 to3 years of age, but may be diagnosed

even later.

Common presentation:

Thalassemia patients who are untreated or inadequately treated have symptoms

such as:

Pale or yellowish skin

Poor feeding, irritability

Abdominal swelling (spleen, liver enlargement)

Infections, fever

Slow growth

Weakness, delayed milestones

Fatigue

Facial bone deformities

The severity of clinical features correlates with the number of and the degree of loss of globin

chains. (Table 2, 3)

Page 136: Training Manual for Hemoglobinopathies and Hemophilia:

136

Table 2

Presenting features of an infant with thalassemia major and cause

Symptom Causative factor Pathogenesis

1. Pallor Severe anemia Switch of hemoglobin chains to . from

fetalhemoglobin(HbF) to adult HbA2

Severity of anemia correlates with number of

functioning globin chains.

Hemoglobin level related to genotype, genetic

modifiers, comorbidities, environmental factors

(e.g., oxidant exposure), and therapy

2 Poor growth Severe anemia

Poor feeding

Feeding problems due to liver and spleen

enlargement and weakness due to anemia

Later growth impairment may be exacerbated by

chronic anemia, effects of extramedullary

hematopoiesis on bones, and endocrine

dysfunction from excessive iron stores affecting

growth and puberty

3 Irritability or

fussiness

Anemia

Often parents feel child is hot, due to anemia

and dyserythropoiesis

4 Hepato-

splenomegaly

Anemia

and Extramedullary

hematopoiesis

The liver and spleen in utero are involved in

hematopoiesis, so they enlarge and try to

compensate for anemia. Trapping of

damaged red blood cells.

Hepatosplenomegaly may be due to a

combination of chronic hemolysis,

extramedullary hematopoiesis in the liver and

spleen, and hepatic iron deposition

5 Fever, may have

recurrent

infections

Infections Anemia, poor nutrition

6 Increased

sweating, child

feels hot (may not

have fever)

Hemolysis

Hemolysis, hyper metabolic state,

Hyperdynamic circulation due to anemia

7 Frontal bossing,

hemolytic facies

Anemia- which is

why it is important

to keep pre-

transfusion

hemoglobin>9g/dl

X-ray hair on end

Expansion of bone due to extramedullary

hematopoiesis

Page 137: Training Manual for Hemoglobinopathies and Hemophilia:

137

Fig 1. Initial work up for suspected patient of Hemoglobinopathy at District hospital/

medical college/ integrated center for Hemoglobinoapthies & Haemophilia (Level 2):

Table 3:

Phenotypic groups of Thalassemia disease –Thalassemia major, intermedia and the

asymptomatic carrier state –thalassemia minor/thalassemia trait

Thalassemia major or

transfusion dependent

thalassemia(TDT)

Thalassemia intermedia

Non transfusion dependent

thalassemia (NTDT)

Thalassemia trait/carrier/

minor

Two genes defective (severe Two genes defective (mild to One gene defect

Page 138: Training Manual for Hemoglobinopathies and Hemophilia:

138

decrease in

beta globin synthesis)

moderatedecrease in beta globin

synthesis)

Severe Pallor requires lifelong

blood transfusions,

irritability,

Hepato-splenomegaly, skeletal

abnormalities,

growth retardation,

Variable degrees of symptoms from

like thalassemia major to

asymptomatic like thalassemia minor,

most are in between.

Have pallor, jaundice,hepato-

splenomegaly, growth retardation,

some requirement of blood

transfusions.

Asymptomatic

Initial Stabilization for a case of suspected thalassemia major or a known case of

thalassemia patient who is ill:

1. The majority of medical management of thalassaemia takes place in the

outpatient clinic and day unit. However, patients occasionally present acutely

and when they do so they may be seriously ill. They need prompt assessment and

management, and will often need admission to hospital.

2. The health care personnel at the level 1 are expected to do the initial stabilization

prior to referral at higher center.

3. If transfusion is indicated in a sick infant and baseline HPLC has not been done,

attempt should be made to preserve the sample for diagnostic hemoglobinopathy

work up prior to transfusion.

Acute presentations may be of a general medical or surgical nature, coincident with the

diagnosis of thalassaemia, but there are also specific clinical presentations which occur

more frequently in thalassaemia and can result in rapid clinical deterioration with

associated morbidity and mortality if not recognised and treated promptly

Evaluation of a sick patient of thalassemia

HISTORY

Specific questions to be asked in history for admitted patient with TM/NTDT :-

i. Infection History

Is the patient feeling unwell, are there rigours, chills, fever? Ask leading questions

for severesystematic illness.

If there are symptoms of infection, consider:

Klebsiella sepsis, which is common in heavily iron-loaded patients. This can result in

septicaemia, urinary tract infection (UTI) or abscesses (cerebral, liver)

Page 139: Training Manual for Hemoglobinopathies and Hemophilia:

139

Yersinia enterocolitica, which may cause abdominal pain, diarrhoea and

lymphadenopathy (more likely with desferrioxamine therapy).

Has the patient been infected with HBV, HCV or HIV?

Thalassaemia patients are vulnerable to malaria infection. Is there malaria or dengue

outbreak in the community?

Does the patient have an indwelling IV device or catheter to provide intensive

chelation whichis a potential source of infection and/or thrombosis.

ii. Splenectomy History

Did the patient undergo splenectomy ?

If so, is he/she taking antibiotic prophylaxis?

(Pneumococcal, meningococcal and haemophilus infections are common in

splenectomised patients).

iii. Transfusion History

If this is the first transfusion, try to do an extended phenotype match. Give 10-

15 ml/kg of PRBC and either prestorageleucodepletedor give transfusion with

a bedside inline leucocyte filter.

Is the patient transfused regularly? If so at what age did this begin?

When was the patient last transfused? Answers may explain Hb levels;

consider the possibility of transfusion-related reactions (short-term or

delayed).

iv. Chelation History

Is the patient taking chelation therapy, and if so, which regime?

Consider the following:

Desferrioxamine- look for sites of sc or iv infusion

-watch for Yersinia and Klebsiellainfection

- ask about changes in eyesight and/or hearing

Deferiprone- check for consequences of reduced neutrophil counts and joint

pains

Deferasirox-investigate renal and hepatic function, -ask about gastrointestinal

disturbances.

Page 140: Training Manual for Hemoglobinopathies and Hemophilia:

140

v. Iron overload History

Adult thalassaemia patients usually know their iron assessment record, including

serum ferritin level and MRI results. Askthefollowing questions.

Has the patient not been adequately chelated recently?

What is the recent trend of serum ferritin? Is it increasing or decreasing?

Has the patient hadan MRI scan or used other tools to assess liver or heart iron

recently?

Does the patient have heart complications?

Is the patient demonstrating decreasing exercise tolerance or breathlessness?

Is there peripheral oedema?

Is the patient experiencing palpitations(awareness of heart beat)?

Are there any symptoms of liver distension with abdominal discomfort?

Does the patient have endocrine complications?

For diabetes, hypogonadism, hypothyroidism and hypoparathyroidism, check

blood and urine sugar, plasma calcium and thyroid function tests.

vi. Surgical history

Has the patient had a splenectomy?

Has the patient had gallstones or cholecystectomy?

vii. Fracture history:

Fractures are common in thalassaemia patients because of weakened bone structure.

Get radiologic evaluation as indicated. Was the patient investigated by DXA scan ?It

is important to know if the patient has a metal plate, which may exclude the

possibility of an MRI examination.

viii Medication history

Is the patient being treated for hepatitis? Many thalassaemia patients are receiving the

current standard for chronic HCV infection and therefore the relevant history needs to

be documented.

Is the patient on anticoagulants? Heart disease and thrombotic conditions (more

common in NTDT) may have necessitated anticoagulant treatment.

Is the patient on antiplatelet drugs?

Page 141: Training Manual for Hemoglobinopathies and Hemophilia:

141

Is the patient on hydroxyurea?

Is the patient on any medication for the heart?e.g. ACE inhibitors, amiodarone,

digoxin

Is the patient on medications for endocrine complications?

e.g. insulin, thyroxin, calcium, vitamin D, estrogens, testosterone,bisphosphonates

EXAMINATION

It is important to categorise the severity of the illness early. Look at the vital signs. Is the

patient in a critical condition?

General appearance

Skin may have a ‘tanned’ discolouration due to iron overload.

Pallor -From anemia.

Jaundice - from anaemia, liver complications or coexistence of Gilbert’s syndrome.

Short stature, short trunk and genu valgum (poor treatment in early life).

Skull and facial changes (signs of TI or poorly treated TM).

Skull enlargement with accentuation of the frontal and malar prominences and

depression of the bridge of the nose. Upper maxillary enlargement affecting teeth

spacing and leading to malocclusion.

Signs of previous surgery – splenectomy, cholecystectomy, fracture surgery.

Pubertal immaturity (as a sign of endocrine failure).

Vital signs

Blood pressure- many TM patients have low BP in steady state. However this may

also be a sign of cardiac decompensation or sepsis.

Pulse - an irregular pulse is an iron-related complication of the myocardium and

patients may be on anticoagulation for this.

Temperature - sepsis is common and life threatening, particularly in iron overloaded

and/or splenectomised patients. Respiratory rate, pulse oximetry, glucose check and

level of consciousness (Glasgow Coma Scale) should be checks of priority.

Hepato-splenomegaly

Most well-treated TM patients will not have had their spleen removed; assessment of

it'ssize may help guide diagnosis of the emergency problem.

Assessment of liver size is also essential. Enlarged liver may result from cirrhosis, right-sided

heart failure or extramedullary hematopoiesis.

Page 142: Training Manual for Hemoglobinopathies and Hemophilia:

142

Bone Marrow Expansion (BME)

BME is increasingly rare in well-transfused TM patients than in TI patients.

Consequences include

- Bone fractures, which are common in both TM and TI and often occur following

trivial injury.

- Micro-fractures –Tenderness in back, ribs, metatarsals due to March fractures of

feet.

- Masses/Weakness due to spinal cord compression

Sepsis -Potential sources/causes

Cholecystitis- Right upper Abdomen tenderness/Lump.

UTI- Renal Angle Tenderness.

Chest- Lung Crepts, Bronchial Breathing.

Teeth-gingivitis, dental abscess, dental cavity.

Endocarditis-Murmurs, Pericardial Rub.

Brain abscess- Papilledema, Focal deficit, Poor GCS.

Meningitis-neck stiffness, poor GCS.

Heart failure

Cardiovascular examination is essential: arrhythmias and biventricular heart failure

from myocardial iron may occur, usually after 10 years of age and can be diagnosed

by irregularities in pulse.

Clinical signs of congestive heart failure, including oedema, require emergency

intervention (iv Desferoxamine ± deferiprone orally).

Pulmonary hypertension is increasingly important, especially in TI with evidence of

heave and loud P2 .

Thrombosis:

Swelling of limb etc or DVT or low saturation hypoxia.

ECG changes of PE.

Endocrine complications

Hypothyroidism,

Hyperparathyroidism and

Diabetes mellitus and other endocrinopathies may be present which may complicate

management and require therapy.

Neurological complications

Extra medullary cord compression.

Brain Abscess and Stroke are common.

Page 143: Training Manual for Hemoglobinopathies and Hemophilia:

143

Look for weakness, focal deficits and evidence of raised intra cranial pressure.

MANAGEMENT

After Diagnosis and stabilization patient should be registered and referred to the day-

care centre for regular management.Routine blood transfusion of filtered PRBC, is the

mainstay of therapy as well as monitoring and appropriate management of iron

overload and evaluation for complications.

Criteria for Inpatient/Emergency management of Thalassemia

Table4:

Sepsis: Fever/suspected infection with Respiratory rate > 22/min or systolic

BP<100mm Hg or GlasGow Coma Scale<15 {qSOFA>2} or any other features of

hemodynamic instability/deranged vital parameters.

Anemia: Worsening anemia despite adequate transfusion or rapid fall from usual

steady state hemoglobin level /persistently increased transfusion requirement.

New onset shortness of breath (dyspnea)

New onset generalized swelling of body /feet (edema/anasarca)

Abdominal pain which is moderate to severe or persistent

Persistent or new onset severe headache

Syncope or altered level of consciousness

New onset focal neurologic deficit /weakness of any limb or face

Acute Back Pain

Trauma with suspected bone fracture or spleen rupture

Any other medical condition where admission is deemed necessary by the treating

physician

Page 144: Training Manual for Hemoglobinopathies and Hemophilia:

144

Causes and symptoms in sick older patients with Thalassemia. major

Symptom Likely Consideration Initial Management Acute

Breathlessness

Severe Anemia

Heart Failure

Pulmonary embolism

Sepsis

Check vitals, oxygen

saturation,

Lung crepitations, irregular

pulse

Get CBC,ECG

Administer oxygen if

required, secure intravenous

line

Consider blood culture, give

first dose intravenous

antibiotic, diuretic prior to

referral

Acute anemia Inadequate transfusion

Folate deficiency

Alloimmunisation

Hemolysis

Parvo virus

Ask for history of transfusion

Look for congestive heart

failure prior to referral.

Fever Sepsis of any focus Check vitals, IV hydration

and antibiotics

Consider baseline cultures

Acute abdominal pain Gallstones with or

without Cholecystitis,

cholangitis,

pancreatitis

Renal stones

Hepatic congestion

from CCF

Intrabdomial infection

Mesenteric/ splenic

infarct or pulmonary

thromboembolism

Check vitals, abdominal

exam

Get ultrasound if available

Secure IV line

Consider analgesia and PPI

prior to referral

Acute Back Pain Osteoporotic Fracture

Spinal Cord Compression due

to extramedullary

hematopoiesis

Page 145: Training Manual for Hemoglobinopathies and Hemophilia:

145

Figure 2: Management of Thalassemia in Outpatient / out patient at Level 2 (District

hospital/ medical college)

Page 146: Training Manual for Hemoglobinopathies and Hemophilia:

146

References

1. Cappellini MD, Cohen A, Porter J, Taher A, Viprakasit V. Guidelines for the Management

of Transfusion Dependent Thalassaemia (TDT). Guidelines for the Management of

Transfusion Dependent Thalassaemia (TDT). Thalassaemia International Federation; 2014.

2. Emergency management thalassemia - Books - NCBI [Internet]. [cited 2019 May 2].

Available from: https://www.ncbi.nlm.nih.gov/books/emergency management thallasemia

3. Taher A, Vichinsky E, Musallam K, Cappellini MD, Viprakasit V, Weatherall SD.

Guidelines for the Management of Non Transfusion Dependent Thalassaemia (NTDT).

Guidelines for the Management of Non Transfusion Dependent Thalassaemia (NTDT).

Thalassaemia International Federation; 2013.

Page 147: Training Manual for Hemoglobinopathies and Hemophilia:

147

Guidelines for Blood Transfusion in Hemoglobinopathies

(Level II- District hospital/ medical colleges )

Thalassemia syndromes can be phenotypically classified into transfusion dependent (TDT)

and non-transfusion dependent thalassemias (NTDT). The former requires lifelong regular

transfusions, while NTDT syndromes affected patients require blood transfusion (BT) only

intermittently & do not depend on transfusions for survival.

‘One size fits all’ approach to transfusion of thalassemia patients, is not applicable. Indeed a

flexible approach crafted to the patient’s individual requirements and to the local availability

of safe blood products is needed for optimal outcomes. Guidelines aim to balance the benefits

of oxygenation and suppression of extra-medullary expansion with those of excessive iron

accumulation from over transfusion.

Goals of Blood Transfusion in Hemoglobinopathy

1. Achievement of appropriate haemoglobin level.

2. Achievement of normal growth & development in paediatric thalassemia patients.

3. Suppression of ineffective erythropoiesis.

4. Prevention of complications splenomegaly , skeletal abnormalities.

5. Minimising complications of transfusion.

Pre-requisite for blood transfusion ( BT )

1. Confirmation of diagnosis ( Mandatory )

2. Testing for HBsAg / HCV / HIV (Mandatory )

3. Vaccination for HBV( Mandatory )

4. Counselling of parents / patients for BT( Mandatory )

Page 148: Training Manual for Hemoglobinopathies and Hemophilia:

148

5. Extended red cell antigen typing that includes at least C, c, D, E, e, and Kell

(if facilities available )

Whom to transfuse?

In newly diagnosed children with thalassaemia, it is very essential to assess the patient

carefully over the first few months after the diagnosis is established and not to embark on

transfusion therapy, too hastily. Emphasize the importance of clinical assessment over

haemoglobin percentage alone. (In most of the cases the diagnosis is made during any

intercurrent illness for which haemoglobin drops from the baseline and the childisbrought for

consultation. ). While the child is under observation—

The Hb level should be checked at least monthly.

Wait till the steady state is reached.

During this period observe the child carefully for growth & activities.

Indication of Blood Transfusion (BT) in Thalassemia major or TDT

patients

Blood transfusion programme to be started if –

Hemoglobin percentage is below 7 g/dl on two occasions 1-2 weeks apart

OR

Any of thefollowing signs & symptoms are present ( Irrespective of haemoglobin

percentage )

Tachycardia

Sweating

Poor feeding

Poor growth

Inability to perform routine activities – attending school

Facial bony changes

Massive splenomegaly

Where possible, this decision should not be delayed until after the 3rd year of age, as the risk

of alloimmunization increases with subsequent difficulty in finding suitable RBC units.

Page 149: Training Manual for Hemoglobinopathies and Hemophilia:

149

Indication of Blood Transfusion (BT ) in NTDT patients

Hemoglobin level should not be a sole determinant of transfusion need except in

patients with considerably severe anemia (hemoglobin level <5 g/dl).

Blood transfusion, if initiated in patients with NTDT will require closer monitoring

and should be individually tailored to meet patient’s needs.

In a patient who never received BT before, if poor growth & development is noted,

start regular transfusions till maximum height is achieved, and bones are fused before

attempting to wean off and then stop regular transfusions.

In case of delayed pubertal growth, child should be on transfusion programme till

pubertal growth is complete.

It is important to maintain the child in good health and normal growth and

development with avoiding unnecessary transfusions in those with minor clinical

features.

Some children with NTDT, specifically with hemoglobin E- βthalassemia, have a

remarkable abilityfor adaptation to low haemoglobin levels and may not require

PRBC(endogenous erythropoiesis). InHbE- thalassemia hemoglobin may be

maintained at lower levels(7g/dl ) as it has beenshown that they do well with lower

Hb%.

Patients having sustained frequent transfusions for extended periods of time should be

managed as per the guidelines for transfusion-dependent β-thalassemia major patients

[28].

Patient should be on regular transfusion in the following settings:

o Falling hemoglobin level in parallel with profound enlargement of the spleen (at a

rate exceeding 3 cm/year in periods of maximal growth and development)

o Growth failure

o Poor school performance

o Diminished exercise tolerance

o Failure of pubertal development

o Signs of bony changes

o Frequent haemolytic crisis (haemoglobin H disease)

o Poor quality of life

Page 150: Training Manual for Hemoglobinopathies and Hemophilia:

150

Occasional blood transfusions should be considered in NTDT patients within any setting

with anticipated acute stress, hemoglobin drop, or blood loss; such as

o Pregnancy

o Surgery

o Infections

Transfusions may be considered for the primary prevention (in high-risk populations), ,

or secondary prevention of the following complications:

o Thrombotic or cerebrovascular disease

o Pulmonary hypertension

o Extramedullary haematopoiesis

o Leg ulcers

Once transfusion therapy is considered, two things need careful monitoring:

o Iron overload

o Risk of alloimmunization.

What is to be transfused?

o Packed Red Blood Cells (Only).

o Hemoglobin concentration of the bag shouldbe minimum 40%.

o Pretransfusion Leukocyte-depleted RBCs(Preferable ).

o If not available use leucocyte filter ( Preferable ).

o ABO and Rh(D) matched blood ( Mandatory ).

o Rh (C, c, E, e) and Kell matching (Preferable ).

o Before each transfusion, perform a full cross-match and screen for new antibodies (

Preferable ).

o Units stored less than 2 weeks (Preferable).

o Washed red cells for patients with repeated severe allergic transfusion reactions or

immunoglobulin A (IgA) deficiency.

In thalassemia major regular blood transfusion is essential and life saving

For any confusion regarding initiation of transfusion programme for thalassemia

intermedia patients, please send the patient to level 3 or 4 for opinion.

Page 151: Training Manual for Hemoglobinopathies and Hemophilia:

151

o Transfusion of blood from first degree relatives should be avoided; this is a bad blood

bank practise.

Transfusion Programme

o Base-line (pre transfusion) haemoglobin % should be maintained at 9-9.5 g% to

suppress hematopoiesis.

o The volume per transfusion is usually 10-15 ml/kg. (Desired – actual Hb) ×

weight × 3 / hematocrit of transfused unit = mL to be transfused.

o Does not waste blood,usual dose 10-15ml PRBC/ kg. Round up to nearest unit

o In uncomplicated patients the whole PRBC unit may be transfused over 1 hour.

o In patients with cardiac failure / very low Hb% e.g. haemoglobin 5 g/dl or below,

give PRBC at a very slow rate,only 1-2 ml/kg/hour.

o The post-transfusion target hemoglobin should be around 12 g/dland should not be

greater than 14 g/dl , though it is not routine practise to check post transfusion HCT.

o A careful record of transfused blood should be maintained for each patient, including

the volume of the administered units, pre-transfusion Hb%, any transfusion reaction.

o The transfusion record must note volume of blood given each time to the patient. If

volume of unit not given by your blood bank, then keep a weighing scale and weigh

bag, and note weight of bag in chart at each transfusion.

o Frequency of blood transfusions - 2-5 weeks depending on need of the child.

o Routine premedication not to be given.

o If patient is having recurrent febrile transfusion reaction – then may give paracetamol

before starting transfusion.

o If the child is in cardiac failure then give Inj Lasix 1 mg / kg before transfusion,

monitor closely during transfusion.

o No need to warm blood bag.

o Do not store blood bag in ward refrigerator.

If there is no increase in hemoglobin after BT on repeated occasion, suspectallo-

antibodies and send the patient to Level 3 or 4 centre for evaluation.

o If requirement of PRBC is> 250 mL / kg / year to maintain baseline haemoglobin 9-

9.5 g/dl, suspect hypersplenism, send to level 3 or 4 center.

Page 152: Training Manual for Hemoglobinopathies and Hemophilia:

152

o A higher target pre-transfusion haemoglobin level of 11-12 g/dl may be appropriate

for patients with heart disease, clinically significant extramedullary hematopoeisis or

other medical conditions

Transfusion Record

Date PreTransfus

ionHb (gm

%)

Volume

transfused

( may put

weight of

bag if

volume

not

known.)

Post

Transfusion

Hb(gm %)

Transfusion

reaction

Remarks

References

1. Sant-Rayn Pasricha, David M. Frazer, Donald K. Bowden and Gregory J. Anderson.

Transfusion suppresses erythropoiesis and increases hepcidin in adult patients with β-

thalassemia major: a longitudinal study .Blood 2013 122:124-133;

2. Cazzola M, Borgna-Pignatti C, Locatelli F, et al. A moderate transfusion regimen may

reduce iron loading in beta- thalassemia major without producing excessive expansion

of erythropoiesis. Transfusion. 1997;37:135–140.

3. Cheng CK, Lee CK, Lin CK. Clinically significant red blood cell antibodies in

chronically transfused patients: a survey of Chinese thalassemia major patients and

literature review. Transfusion. 2010;52:2220–2224

4. Michail-Merianou V, Pamphili-Panousopoulou L, Piperi-Lowes L, et al.

Alloimmunization to red cell antigens in thalassemia: comparative study of usual

versus better-match transfusion programmes. Vox Sang. 1987;52:95–98.

5. Cappellini MD, Cohen A, Porter J, Taher A, Viprakasit V. Guidelines

fortheManagementofTransfusionDependentThalassaemia(TDT).Ed 3. Nicosia,

Cyprus: Thalassaemia International Federation; 2014.

Page 153: Training Manual for Hemoglobinopathies and Hemophilia:

153

6. VichinskyE, NeumayrL, TrimbleS, etal.Transfusioncomplicationsin thalassemia

patients: a report from the Centers for Disease Control and Prevention (CME).

Transfusion. 2014;54:972-981; quiz 1.

7. .Garbowski MV, et al.Residual erythropoiesis protects against myocardial

hemosiderosis in transfusion-dependent thalassemia by lowering labile plasma iron

via transient generation of apotransferrinHaematologica 102(10) 1640-1649 2017).

8. How I treat thalassemia Eliezer A, Patricia J. Blood. 2011;118:3479-88

9. Challenges of alloimmunization in patients with haemoglobinopathies Chou ST, Liem

RI, Thompson AA British journal of haematology. 2012;159(4):394-404.

10. Standards of care guidelines for thalassemia. Children’s Hospital & Research Center

Oakland; 2012 (http://thalassemia.com/treatmentguidelines-4.aspx). Accessed April

20, 2015.

11. Guidelines for the management of non transfusion dependent thalassaemia (NTDT):

Thalassaemia Taher A, Vichinsky E, Musallam K, Cappellini M-D, Viprakasit

V. International Federation, Nicosia, Cyprus; 2013.

12. Rivella S. The role of ineffective erythropoiesis in non-transfusion-dependent

thalassemia. Blood Rev 2012;26 Suppl1:S12-15.

13. Allen A, Fisher C, Premawardhena A, peto T, Allen S, Arambepola m, Thayalsutha

V, olivieri N, Weatherall D. Adaptation to anemia in hemoglobin E-ss thalassemia.

Blood 2010;116(24):5368-5370.

Page 154: Training Manual for Hemoglobinopathies and Hemophilia:

154

Guidelines for Chelation therapy in Thalassemia

At level 2 centers

(Comprehensive ICHH centers for hemoglobinopathies and hemophilia)

Iron overload is the main cause of morbidity and mortality is hemoglobinopathies.Iron

overload occurs when iron intake is increased over a sustained period of time, either as a

result of red blood cell transfusions in transfusion dependent thalassemia (TDT) or

thalassemia major or increased absorption of iron through the gastrointestinal (GI) tract in

non transfusion dependent thalassemia (NTDT) or thalassemia intermedia. Proper monitoring

of iron overload and chelation therapy is a very important component of treatment of

thalassemia patients. Chelation therapy aims to balance the rate of iron accumulation by

increasing iron excretion in urine and or faeces. The major challenge in chelation therapy is

to achieve regular adherence to treatment regimens. The goal is to equip the ICHH centers at

District hospitals or medical colleges to screen, and start iron chelation. Complicated cases or

non responders who need more evaluation should be referred to level 3 (COE) or 4 (National

referral center) for optimal investigation and care.

Monitoring of Iron Overload

Available methods-

Serum ferritin

Advantages

• Easy to assess

• Inexpensive

• Repeat serial measures are useful for monitoring chelation therapy

• Positive correlation with morbidity and mortality

Page 155: Training Manual for Hemoglobinopathies and Hemophilia:

155

Disadvantages

• Indirect measurement of iron burden

• Fluctuates in response to inflammation, abnormal liver function

• Most serum ferritin assays were developed mainly for detecting iron deficiency, and

the linear range of the assay at high SF values needs to be known. Need to dilute

samples with high values, to give readings within the linear range of the assay

• May not provide reliable indication of iron levels in cases of non transfusion

dependent thalassemia (NTDT) or thalassemia intermedia.

Liver iron concentration (LIC) measurement

Gold standard for iron assessment- liver biopsy

Methods – by liver biopsy

Advantages

• Direct measurement of LIC

• Quantitative, specific, and sensitive

• Provides information on liver histology/pathology

• Positive correlation with morbidity and mortality

Disadvantages

• Invasive, painful, potentially serious complications, eg, bleeding

• Practically not a feasible option.

Liver iron concentration (LIC) by T2* MRI

Advantages

• Assesses iron content throughout the liver

• Status of liver and heart can be assessed in parallel

• Validated relationship with LIC

• Allows longitudinal patient follow-up

Disadvantages

• Indirect measurement of LIC

• Requires MRI imager with dedicated imaging method,ensure the lab has proper

validation and standardization.

• Children younger than age 7 years may require a general anaesthetic

Page 156: Training Manual for Hemoglobinopathies and Hemophilia:

156

Cardiac iron by T2* MRI

Advantage

• Non-invasive

• Reproducible

• Have predictive value in identifying patients at high risk of developing

deterioration in LVEF

Disadvantages

• Indirect method

• Non-linear relationship with myocardial iron content ( MIC )

• Technically demanding ensure the lab has proper validation and

standardization.

Recommendations

Recommended method for monitoring iron overload is Ferritin ( at level 2)

Monitoring for Ferritin to be started after 2 years of age or after 10-20 BT in cases of

TDT

Monitoring to be started for all NTDT patients >10 years of age.

All patients may be screened by T2*MRI after age of >10 years annually or every two

years, they can be referred to the State center facility for this test.

T2* MRI for LIC to be done in NTDT patients with features of iron overload &

disproportionately low ferritin – patient to be referred to level 3 or 4

T2*MRI for MIC in patients with high Ferritin value or deteriorating cardiac function

– patient to be referred to level 3 or 4.

When to start chelation therapy?

Transfusion dependent thalassemia( TDT )

2 years of age or older,

1 to 2 years of chronic transfusions,

Serum ferritin level > 1000 ng/mL on 2 separate measurement obtained when the

child is well.

LIC of at least 7 mg/g dry weight( If available )

TDT patients < 2 years of age requiring chelation therapy to be referred to level 3 or

4 for opinion.

Page 157: Training Manual for Hemoglobinopathies and Hemophilia:

157

Non Transfusion Dependent Thalassaemia( NTDT )

More than 10 years of age orSerum ferritin level ≥750 ng / ml.

Liver iron concentration ≥5 mg Fe/g dry weight ( If available )

Goal of Chelation Therapy

To prevent the toxic effects of iron overload

To avoid chelator toxicities

To maximize adherence to therapy

To prevent organ dysfunction

o Liver iron concentration (LIC) <5mg/g dry weight,

o Cardiac T2* MRI >20ms

Recommendations

Chelation therapy to be started

1. TDT if Ferritin > 1000 ng / mL & the child is> 2 years old.

2. NTDT if Ferritin > 800 ng / mL & the child is 10 years of age

What Chelating agent to start?

Available iron chelation drugs

1. Deferroxamine (DFO)

2. Deferiprone (DFP)

3. Deferasirox (DFX)

Deferasirox – oral drug ( 1st line drug )

Thalassemia major Dose : 20 mg / kg/day, increase dose by 5-10 mg / kg every 3 months

based on Ferritin report ( see table 5, 6)

Dose in NTDT-10 mg/k/day

Escalation of dose in NTDT

At 1 month

No escalation if Ferritin < 1500 ng / mL ( Baseline )

To 15 mg /kg / day if Ferritin > 1500 < 3000 ng / ml, ( Baseline )

Page 158: Training Manual for Hemoglobinopathies and Hemophilia:

158

To 20 mg /kg/day if Ferritin > 3000 ng / mL ( Baseline )

At 6 months

Ferritin < 1500 ng / mL – no escalation

Ferritin > 1500 < 3000 ng / mL – escalate by 5 mg / kg/day ( maximum 20 mg / kg / day )

Ferritin > 3000 ng / mL – escalate by 10-15 mg / kg / day ( maximum 30mg / kg / day ) :

refer to level 3 or4

DFX is administration on empty stomach dissolved in a glass of water /fruitjuice, do

not use any metalic glass /spoon.

DFX can be given in two divided doses if the patient cannot tolerate once daily dose.

Twice daily dose is equally effective

Contraindicated: in patients with renal failure or significant renal dysfunction (see

below).

Caution is recommended for patients with advanced liver disease and hepatic

decompensation

Deferasirox to be discontinued if Ferritin < 300 ng / ml, but continue monitoring so

that it can be restarted as the ferritin will increase.

Table 5:

Monitoring for Deferasirox

Time Parameters

Before starting Deferasirox

Serum ferritin

Serum creatinine x2

Liver function tests

1st month of treatment Serum creatinine (weekly )

Monthly

Serum creatinine

Liver function tests ( every two weeks for first month )

3 monthly

Serum Ferritin

Annual Hearing and vision check up

Deferiprone : oral drug ( 2nd line drug )

Dose 75 mg/kg/day, given in three divided doses

Should not be started in patient < 6 years of age

Require monitoring of blood count

Page 159: Training Manual for Hemoglobinopathies and Hemophilia:

159

Each patient’s absolute neutrophil count should be measured before starting DFP

therapy and weekly during treatment. Should be stopped if ANC < 1500. Refer to

level 3 or 4

Deferiprone is not recommended in HIV positive or in immunocompromised patients,

due to its risk of agranulocytosis.

Arthropathy range from mild pain to severe arthritis, spontaneous recovery occurs

despite continuing therapy. If joint symptoms continue despite , reduce dose of

DFP, if not controlled after dose reduction , stop DFP. Refer to level 3 or 4

Increased ALT values usually asymptomatic and transient and ALT usually

returns to normal without discontinuation or dose decrease.

Deferiprone is renally excreted so there may be an increased risk of complications in

patients with impaired renal function

Desferrioxamine( DFO ) – 3rd line drug

Delivery: Subcutaneous or IV infusions

Dosage: 30-60mg/kg/day 12hours subcutaneous infusion 5-7 times per week

Average dose should not exceeds 40 mg / kg/ day until growth has ceased

IV dose only in severely iron over loaded patients with cardiac failure .

Needs special infusion pump

Disadvantages

Expensive, time-consuming, not comfortable

Local skin reactions

Infection with Yersinia enterocolitica

Severe hypersensitivity reaction

Hearing problems

Effects on the eye

Growth retardation

Skeletal changes

Page 160: Training Manual for Hemoglobinopathies and Hemophilia:

160

Table6:

Dose adjustment in transfusion dependent thalassemia (TDT) or thalassemia major

LIC( preferable, may

refer)

Serum Ferritin

( Mandatory )

Recommendation

< 3 mg / g < 1000 ng/ mL

Lower dose if Ferritin < 1000 ng / ml

Stop if < 500 ng / ml

3 –7 mg / g 1000- 2500 ng / ml

Continue existing therapy

> 7mg / g > 2500 ng / ml Increase dose

T2* < 20 ms without

cardiac dysfunction

If planning to Start Desferioxamine

Refer to level 3 or4

T2* < 20 ms with

cardiomyopathy or T2* <

10 ms without

cardiomyopathy

Before Combination therapy ( DFO

+ DFP )

Refer to level 3or4

Management of toxicities related to iron chelation medicines

1. Gastrointestinal events are relatively frequent with DFX therapy but are typically mild

to moderate and include diarrhoea, abdominal pain, nausea and vomiting, occurring in

approximately 15-26% of patients. These symptoms rarely require dose adjustment or

discontinuation, and decrease year on year over 5 years of follow up. Can be given in

two divided doses if gastrointestinal problem persists. – Refer to level 3 or 4 for

opinion

2. Rash typically develops within two weeks of starting treatment. Mild rashes often

resolve without dose modification, and became very rare after year 1 of treatment. For

moderate to severe rashes, treatment should be stopped and later restarted at a very

low dose (<5 mg/kg). Severe skin rash associated with angioedema is rare, and DFX

therapy may need to be halted completely --Refer to level 3 or 4.

3. Renal dysfunction- in cases of renal dysfunction the dose can be reduced by 10

mg/kg, if serum creatinine increases >33% above pretreatment measurements on two

Page 161: Training Manual for Hemoglobinopathies and Hemophilia:

161

consecutive visits and no other cause is found. Refer to level 3 or 4 for opinion as

thalassemia major patients can have renal dysfunction due to their disease..

4. Liver dysfunction- ALT if increased, chelation should be stopped & reintroduced at

low doses once ALT is normal. Refer to level 3 or 4 for opinion.

Recommendations

1. Recommended 1st line therapy is Deferasirox.

2. Chelation to be started after 2 years of age & ferritin > 1000 ng / mL in TDT &> 800

ng / ml& 10 years of age in NTDT.

3. Deferasirox can be given with food,not required to be on an empty stomach once or

twice a day regularly and daily.

4. Shift to twice daily dose of Deferasirox in cases of persistent gastrointestinal

symptoms.

5. In case of severe toxicity of DFX , DFP ( 2nd

line drug ) is recommended provided the

child is > 6 years old

6. In case of persistent or severe adverse effects patients to be referred to higher level

( COE ) or level 4 centerfor opinion

7. Before starting 2nd

or 3rd

line drug, patient to be referred to level 3 (COE) or

National level referral center (level 4 )for opinion.

8. Desferrioxamine IV injection require placement of a central venous catheter for

continuous infusion, in case of emergency it can be given through peripehral vein

diluted in at least 100ml of normal saline.

Page 162: Training Manual for Hemoglobinopathies and Hemophilia:

162

Table7:Summary of chelating agents

Category DFO

(DESFERRIOXAMINE)

DFP

(DEFERIPRONE)

DFX

(DEFERASIROX)

Children age

2-6

May be used AVOID First line

Children age

> 6 and

adults

May be used 2nd

line First line

Route s.c. / i.m. or i.v injection Oral, tablet or liquid Oral, dispersed tablet

Dosage and

frequency

20 –64 mg/kg 5 -7 × / week,

50 mg/kg in EU

Children’s dose up to 40

mg/kg

75 -100 mg/kg/day

in 3 divided doses

daily

20-40 mg/kg/day once

daily.

Lower doses in NTDT

Contra-

indications

- Pregnancy

- Hypersensitivity

- Pregnancy

- History of

neutropenia or

condition with

underlying risk of

cytopenia

-

- Pregnancy

- Hypersensitivity

- Hepatic impairment

or renal failure

Precautions Monitor audiometry

regularly

- Monitor eyes regularly

- Fever suggestive of

septicemia with organisms

that use ferrioxamine

(Yersinia, klebsiella)

- Measure

neutrophil count

(ANC) before

starting and monitor

ANC weekly

- For neutropenia :

ANC < 1.5 × 109/L)

interrupt treatment

- For

agranulocytosis

(ANC < 0.5 × 109/L),

consider

hospitalization

- Monitor for

symptoms of

arthropathy

- Monitor liver

function regularly

- Monitor creatinine

monthly

-

- Monitor liver

function regularly

Page 163: Training Manual for Hemoglobinopathies and Hemophilia:

163

NOTES

If any patient wants to conceive, chelation therapy should be stopped 3 months

before.

Adjunctive Vitamin C is not recommended.

I.V DFO along with blood transfusion not recommended.

IV DFO is recommended in continuous drip in patient with cardiac failure only(

Patient to be referred to level 3 or 4 for opinion before this )

Combination therapy

If a patient is failing on first line therapy, dose adjustment and attention to adherence

(practical as well as psychological support) are the next steps. If this fails then regime

adjustment can be considered, depending on the circumstances.

Failing this a combination therapy is recommended. Patient may bereferred to level 3

or 4 before startingthe combination therapy.

Following are the recommended combinations

Combined DFO and DFP : DFO given twice a week ( Week end ) combined with

DFP daily at standard doses (75 mg/kg/day).

DFX 20-40 mg/kg/d 7 days per week, plus DFO 40 mg/kg/d 5 days per week

Target of Iron chelation

Ferritin levels between< 500 ng/mL in TDT

Ferritin level < 300 ng / mL in NTDT

LIC - < 5 mg/g dry weight NTDT

LIC < 7 mg / g dry weight TDT

Cardiac T2* >20 ms.

Monitoring chelation therapy (seeTable7, 8)

Monthly biochemistry (creatinine and liver function tests) ( Mandatory )

3 monthly Serum ferritin (Ferritin levels may be inconsistent, especially in sickle ce

ll patients, so decisions must be based on trends over time) ( Mandatory )

Cardiac T2* MRI annually in selected patients (Refer to higher center)

Page 164: Training Manual for Hemoglobinopathies and Hemophilia:

164

Once yearly Ferriscan of liver in selected patients ( Optional )

Annual audiometry and ophthalmology review ( Mandatory )

Additionally, patients on Deferiprone require careful monitoring of neutrophil count

preferably weekly at commencement of therapy ( Mandatory )

Local reactions with DFO - Persistent local reactions may be reduced by varying

injection sites, lowering the strength of infusion, or in severe cases, by adding 5-10

mg of hydrocortisone to the infusion mixture. Application of topical low potency

corticosteroid cream after injection can reduce local reactions.

Table 8:Monitoring chelation

DFO

DFX DFP

ANC *

(absolute neutrophil

count)

- - Weekly for 1 month

& then monthly

Liver function test

(LFT)

- Every month Every 3 months

Creatinine Every 3 months Every month Every 3 months

Zinc, Copper ,

Calcium,

Magnesium

Annually Annually Annually

Eye Examination Annually Annually Annually

Audiogram Annually Annually Annually

Sitting height Biannually Biannually Biannually

Clinical monitoring Every month Every month Every month

T2 *MRI

heart / Liver

After 10 years

Every 2 years

After 10 years

Every 2 years

After 10 years

Every 2 years

* Absolute neutrophil count is Calculated by formula-

total WBC count xtotal neutrophils (segmented neutrophilis%+band %) x 10=ANC

Page 165: Training Manual for Hemoglobinopathies and Hemophilia:

165

CONDITIONS WHEN PATIENT TO BE REFERRED TO LEVEL 3 OR 4

1. NTDT patients with features of iron overload & disproportionately low ferritin –

patient to be referred to level 3 or forT2* MRI for LIC.

2. Patients with high Ferritin value & deteriorating cardiac function – patient to be

referred to level 3 or 4 centersfor T2*MRI for MIC .

3. TDT patients < 2 years of age requiring chelation therapy to be referred to level 3

(COE) or 4 (National referral center) for opinion.

4. For any patient with chelation failure

5. For starting 2nd

or 3rd

line chelating agent

6. For starting combination of chelating agents

7. For persistent toxicity of chelating agent

References:

1. Adamkiewicz TV, Abboud MR, Paley C, et al. Serum ferritin level changes in children with

sickle cell disease on chronic blood transfusion are nonlinear and are associated with iron load

and liver injury. Blood. 2009;114:4632–4638.

2. Anderson LJ, Holden S, Davis B, et al. Cardiovascular T2-star (T2*) magnetic resonance for the

early diagnosis of myocardial iron overload. Eur Heart J. 2001;22:2171–2179]

3. Kirk P, Roughton M, Porter JB, et al. Cardiac T2* magnetic resonance for prediction of cardiac

complications in thalassemia major. Circulation. 2009b;120:1961–1968.

4. Piga A, Galanello R, Forni GL, et al. Randomized phase II trial of deferasirox (Exjade, ICL670),

a once-daily, orally-administered iron chelator, in comparison to deferoxamine in thalassemia

patients with transfusional iron overload. Haematologica. 2006;91:873–880.

5. Pongtanakul B, Viprakasit V. Twice daily deferasirox significantly improves clinical efficacy in

transfusion dependent thalassaemias who were inadequate responders to standard once daily dose.

Blood Cells Mol Dis. 2013;51:96–97.

6. Angelucci E, Brittenham GM, McLaren CE, et al. Hepatic iron concentration and total body iron

stores in thalassemia major. N Engl J Med. 2000;343:327–331.

7. Aydinok Y, Kattamis A, Cappellini MD, et al. Deferasirox– Deferoxamine Combination Therapy

Reduces Cardiac Iron With Rapid Liver Iron Removal In Patients With Severe Transfusional Iron

Overload (HYPERION)(abstract). Blood. 2013:2257.

8. Olivieri NF, Buncic JR, Chew E, et al. Visual and auditory neurotoxicity in patients receiving

subcutaneous deferoxamine infusions. N Engl J Med. 1986;314:869–873.

Page 166: Training Manual for Hemoglobinopathies and Hemophilia:

166

9. Aydinok Y, Ulger Z, Nart D, et al. A randomized controlled 1-year study of daily deferiprone plus

twice weekly desferrioxamine compared with daily deferiprone monotherapy in patients with

thalassemia major. Haematologica. 2007;92:1599–1606.

10. Taher AT, Porter JB, Viprakasit V, et al. Deferasirox effectively reduces iron overload in non-

transfusion-dependent thalassemia (NTDT) patients: 1-year extension results from the

THALASSA study. Annals of hematology. 2013;92:1485–1493. Cappellini MD, Bejaoui M,

Agaoglu L, et al. Iron chelation with deferasirox in adult and pediatric patients with thalassemia

major: efficacy and safety during 5 years’ follow-up. Blood. 2011;118:884–893.

11. Otto-Duessel M, Aguilar M, Nick H, et al. Comparison of twice-daily vs once-daily deferasirox

dosing in a gerbil model of iron cardiomyopathy. Exp Hematol. 2007;35:1069–1073.

12. Ceci A, Baiardi P, Felisi M, et al. The safety and effectiveness of deferiprone in a large-scale, 3-

year study in Italian patients. British journal of haematology. 2002;118:330–336.

13. Chiodo AA, Alberti PW, Sher GD, et al. Desferrioxamine ototoxicity in an adult transfusion-

dependent population. J Otolaryngol. 1997;26:116–122.

14. Cohen AR, Galanello R, Piga A, et al. Safety and effectiveness of long-term therapy with the oral

iron chelator deferiprone. Blood. 2003;102:1583–1587.]

15. Cohen AR, Mizanin J, Schwartz E. Rapid removal of excessive iron with daily, high-dose

intravenous chelation therapy. J Pediatr. 1989;115:151–155.

16. De Virgillis S, Congia M, Frau F, et al. Desferrioxamine-induced growth retardation in patients

with thalassaemia major. Journal of Pediatrics. 1988;113:661–669.

Page 167: Training Manual for Hemoglobinopathies and Hemophilia:

167

Monitoring and data collection

Routine Outpatient management of Thalassemia Major at level 2

All patients of Thalassemia need to follow up every month and monitored as per the

monitoring section 4, with the dedicated Thalassemia care doctor, nurses and counselor. This

must be supervised by a ICHH centre in charge who can be the pediatrician /medical officer

in charge of the ICHH, or Medical superintendent. This person is responsible for medical

care, record keeping and provision of transfusion, medicines, monitoring and timely referral.

The emphasis is on safe blood transfusion, monitoring of iron overload and documentation.

Register in hemoglobinopathy registry.

Children with thalassemia major or NTBT on regular blood transfusion and chelation

therapyrequire regular monitoring.

Medical staff at Hemoglobinopathy Center should maintain the records properly at regular

intervals as indicated on Table I – IV. Children need to be referred to higher center for tests

which can’t be undertaken at day care center. It is the responsibility of the staff of day care

center to get the appointment for tests or blood samples may to be sent to higher center in a

proper transport system for detailed investigation.

1. Adequacy of Blood transfusion therapy with packed red blood cells (pRBCs)

2. Monitoring for adequacy of Iron chelation for iron overload and adverse effects of

drugs

Page 168: Training Manual for Hemoglobinopathies and Hemophilia:

168

3. Monitoring of complications due to the disease and their treatment with special

emphasis on growth

4. Management of endocrine complications (endocrine, cardiac, skeletal etc.),

5. Discussion of Bone marrow transplantation (BMT)/ Hematopoietic stem cell

transplant (HSCT) if considered

6. Psychological support.

7. Growth assessment is important and for monitoring accurate maintenance of records

are necessary. Height & weight of all these children needs to be recorded on IAP

growth charts (Fig 3&4) to ensure normal growth velocity. Showing of growth

velocity is an early indication of growth retardation.

Integrated centres for hemoglobinopathies and hemophila.

Ensure all hemoglobinopathy patientsare enrolled in Registry as soon as

diagnosis is made.

Enroll at ICHH center for regular care and monitoring, get patient address,

contact number.

Make sure counselling of family for disease and complications is completed.

Make sure Genetic counselling is provided to family. Encourage cascade testing

of family members.

Thalassemia major patients are on regular blood transfusion and chelation

therapy so they need regular monitoring to see for efficacy of chelation, any

modification if needed and to evaluate for toxicities and complications.

Usually the monitoring and data collection is to be done at the integrated center

for haemophilia and hemoglobinopathies attached to Pediatrics/

Hematologydepartment in Medical college or District hospital.

Routine information to be captured for Hemoglobinopathies

Demographic-Complete name address, sex age, diagnosis

Contact number

Baseline tests- blood grouping extended, other important issues

Page 169: Training Manual for Hemoglobinopathies and Hemophilia:

169

Table 9:Monitoring at each visit/transfusion

Name, age, address, hospital number

Date Date Date

Weight

Pre transfusion hemoglobin

Note :- Routine post – transfusion Hb checking is not

recommended.

Liver size (by clinical examination)

Spleen size (by clinical examination)

Volume of PRBC given

Transfusion reaction

Immunizations up to date?

Current medications

1.Folic Acid

2.

3

Table 10:Monthly monitoring for patients

Date Date Date

If on deferasirox – SGPT, SGOT, BUN, S.creatinine, Urine

R/E

If on deferiprone, hydroxyurea

– Complete blood counts including DLC

Next date of transfusion

For SCD/T intermedia

Hydoxyurea

For sickle cell disease/ post splenectomy

Penicillin prophylaxis Other immunizations given

(Pneumococcal etc)

Table 11:Monitoring every 6 months for all patients

Date Date Date

S. ferritin (ng/ml)

SGPT

SGOT

BUN

S. Creatinine

Calcium

Phosphorus

Height

Weight

Growth velocity

Page 170: Training Manual for Hemoglobinopathies and Hemophilia:

170

Table 12: Monitoring every year

Date Date Date

Anti HBs antibody

HCV I gG antibody

HIV 1 & 2

The following tests to be performed annually after the age of 10 years

Blood sugar ( fasting) or GTT

TSH

ECG

Echocardiography

MRI T2* Heart/Liver (for all patients with iron overload)

DEXA Scan

Other endocrine work up as per Level 3 or 4 consultation

Additional tests for SICKLE CELL DISEASE PATIENTS ( SCD)

2-18 years :Trans cranial Doppler

>6 years : URINE Micro Albumin

>6 years : Pulmonary function test

While most of the above investigations for monitoring are available at district

hospitals or state medical colleges, patients may be referred to Institutes of National

importance for annual T2*MRI Heart/Liver at following centers:-

A few centers where these tests are standardized and available are given below. This

list will need updating periodically.

AIIMS, New Delhi

PGIMER, Chandiagarh

CMC,Vellore

SGPGI, Lucknow

Sri Chitra Tirunal for Medical Science and Technology, Thiruvananthapuram

Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bangalore.

Children <10 years with Thalassemia do not routinely requireannual Dexa Scan and

T2*MRI Heart/Liver

Page 171: Training Manual for Hemoglobinopathies and Hemophilia:

171

Fig 3, IAP Boys growth chart

Page 172: Training Manual for Hemoglobinopathies and Hemophilia:

172

Fig 4, IAP Girls growth chart

Page 173: Training Manual for Hemoglobinopathies and Hemophilia:

173

Common Complications and their management in

Thalassemia major

Guidelines for management of complications and long term follow up at level 2/3

Contents

Alloimmunization and its management

Hypersplenism

Transfusion related infections

Cardiac complications

Hepatic complications

Endocrine complications

Indications for splenectomy

Long term follow up and monitoring

One must be aware, that despite adequate iron chelation therapy most of the patients will

develop complications. This is because of excessive iron which gets deposited in different

organs like heart, liver and various endocrine glands which results in their dysfunction /

toxicity of these organ systems. Iron deposition in various endocrine glands may cause

hypothyroidism, hypoparathyrodism, diabetes, growth and development retardation, bone

demineralization and skeletal deformities. Iron chelation if started appropriately and well

monitored for efficacy will reduce the severity of complications.

Patients are also at higher risk to develop transfusion transmitted infections such as

Hepatitis B, C or HIV.

These children need to be monitored for toxicity of iron chelation therapy. Therefore,

management of Thalassemia needsa multidisciplinary team approach consisting of

paediatrician, cardiologist, gastroenterologist, endocrinologist and many problems will

require a haematologist, if not already supervising.

Page 174: Training Manual for Hemoglobinopathies and Hemophilia:

174

There may be limitations of staff & facilities to investigate and diagnose various

complications. However medical staffat the integrated hemoglobinopathy centersshould

have adequate information & knowledge for earlysuspicionof these complications. Then

they need to get the tests performed or refer or make prompt arrangement for appropriate

evaluation at level 2 for their management.

A. Difficulty in maintaining Hb above 10 gm% despite adequate blood

transfusion

The common factors for inability to maintain pre transfusion average hemoglobin of

10g/dlshould be excluded (Table 13). Higher transfusion therapy usually decreases the spleen

enlargement & effect of hypersplenism. Thereis evidence that average transfusion

requirement is 20–30% higher in unsplenectomised than splenectomised thalassemia major

patients, but due to complications splenectomy is to be avoided, unless the hypersplenism is

causing serious compromise and higher transfusion is not manageable by chelation therapy.

1) Alloimmunization–

Children with thalassemia are at higher risk of developing allo-antibodies as a result of

mismatch of minor blood groups. The prevalence of allo-immunization in various counties

varies between 3 to 37 % in various studies. However its prevalence in our country varies

between 3.7 to 15.5%.

Alloimmunization occurs more frequently if blood transfusions are initiated between 18-24

months of age in thalassemia major (TM) or NTBT patients; after splenectomy and in those

who are not getting leucodepleted blood transfusion. Developmentofalloimmunization in

TDT can be prevented by (a) initiation of leucodepleted blood transfusion from the beginning

(b) extended blood group matching at the beginning and by providing proper cross matched

blood against major and minor blood group antigens from the beginning (c) by avoiding

splenectomy.

Development of alloantibodies should be suspected at the ICHH if -

(a) Child develops jaundice within 48 hours of blood transfusion

(b) Passing of high coloured urine

(c) Increased blood transfusion requirement which can be detected by maintaining

accurate amount of blood transfused every time

(d) Difficulty in maintaining the Hb levels

(e) Difficulty in cross matching

All these children need to be referred to level 4 or level 3 centers where the following

facilities are available.

(I) Antibody screening

(II) Identification of antibodies with adequate cell line panel

Page 175: Training Manual for Hemoglobinopathies and Hemophilia:

175

(III) Facilities for extended blood group match.

Children with allo-immunization need to be managed at level 3 or 4 centers. However blood

can be transfused if appropriate cross matched blood is made available from higher centerto

the ICHH centers. (Integrated centre for Hemoglobinopathies & Haemophilia )

Table 13:

Causes of Low Hemoglobin levels

2) Hypersplenism

This complication may be preventable if adequate transfusion is initiated from the beginning

and average pre-transfusion hemoglobin is maintained between 9.5 to 10.5g/dl. It can be

suspected if spleen size is gradually increasing, this can be easily detected in integrated

hemoglobinopathy center by recording the spleen size correctly during every visit. Blood

transfusion requirement exceeding 200ml/kg/year is another indication of hypersplenism.

In the day care center,amount of blood transfused at each visit should be recorded & average

requirement over the last 6 months needsto be calculated periodically at 6 months interval for

its early detection. Development of mono or bicytopenia further supports the possibility of

development of hypersplenism. Such children need to be referred to level 2 for further

evaluation & advice.

These children should be administered vaccines for pneumococcal, meningococcal & HIB

vaccine atleast 3 weeks prior to splenectomy. Children need to undergo splenectomy at level

2 or higher centers. Surgeon may decide laparoscopic or open splenectomy based upon on the

size of spleen and other related factors. All these children need to be given Penicillin

prophylaxis preferably life-long or at least till the age of 20 years.

General Poor Education of Parents & Patients Long Distance from House – DCC Doctors transfused blood only when Hb< 8 gm/dl Only one unit of blood transfused irrespective of Wt / Hb Poverty

Social Belief maintaining high Hb – Iron Overload Family functions (Marriages / House problems ) School exams Summer vacations Family Trips

Page 176: Training Manual for Hemoglobinopathies and Hemophilia:

176

Table 14:Main indications for splenectomy in current era, the medical practice is to

avoid unless necessary:

Indications Comment

Increased blood requirement that

prevents adequate control with iron

chelation therapy

Annual transfusion volume

(75% hematocrit) used to flag

an increased blood requirement

(200–220 ml/kg/year)

Alloimmunization, concurrent

infections, suboptimal

transfusion therapy should be

ruled out

Hypersplenism

Cytopenias

Symptomatic splenomegaly Accompanied by symptoms

such as left upper quadrant

pain or early satiety

Massive splenomegaly causes

concern about possible splenic

rupture.

Post Splenectomy Sepsis

These children & their parents should be counselled as these children are at higher risk of

developing sepsis. Parents should be advised to start Amoxicillin / Septran at the onset of

fever at home. They should take the child preferably at level 2centers otherwise to integrated

center for hemoglobinopathy, or emergency (centers should develop protocols for their

hospital to reduce time before being seen by a doctor) for immediate admission & treatment.

In these children blood culture should be taken, blood for malarial parasite on peripheral

blood smear and blood malaria antigen should be undertaken along with Xray of chest to rule

out pneumonia etc. These children need to be treated with broad spectrum antibiotics

providing adequate cover for gram positive & negative organism. Choice of antibiotics

should be dependent on Institutional policy or with third generation cephalosporin such as

cefotaxime or ceftriaxone with vancomycin.

Page 177: Training Manual for Hemoglobinopathies and Hemophilia:

177

Early detection of TTI

Development of hepatitis B or C and HIV infection can be detected only by screening these

children for these infections at yearly interval. Children who are found to be positive need to

be referred to level 3 or 4 centers for detailed evaluation & appropriate management.

B) Transfusion related infections

Hepatitis C virus (HCV), hepatitis B virus (HBV), human immunodeficiency virus (HIV)

and syphilis are the infectious agents usually transmitted via packed red blood cell

(pRBC) transfusions.

Physicians should be aware of the potential life threatening infections in thalassemic

patients. All family needs to be counselled about these infections. Besides these common

TTI other infections which can also be transmitted following blood transfusions include

Cytomegalovirus (CMV), Parvo-B-19, West Nile Virus (WNV), Dengue etc.

Use of storage leucodepleted RBCs stored <14 days reduce risk of CMV, herpes viruses

(e.g Epstein Bar Virus) human T cell lymphotropic virus type I, Yersinia and protozoa

infections (Leishmania species &Trypanosomacruzi)

All patients with TDT should be protected by vaccination against HBV. As the protection

offered by vaccination is not absolute & it wears off over time. Therefore these children

should be given booster vaccine regularly at 5 years interval.

Almost these TTI, malaria may manifest with feverwhich may be associated with rigor

&chills. If any child develops fever within 7-10 days of blood transfusion. They need to

be investigated for TTI & should be treated at least at a level 2center.

C) Cardiaccomplications Cardiac complications secondary to iron overload remain the major cause of morbidity &

mortality. Heart failure is the major complication due to iron accumulation incardiac muscles.

However cardiac complications have been divided in two major groups (Fig. 5).

1) Iron Overload Complications

a) Reversible congestive heart failure

b) Arrhythmias, heart blocks

2) Non iron overload complication

a) Pulmonary hypertension

b) Arrhythmias such as atrial fibrillation (AF)

c) Thrombotic strokes related to AF

d) Cardiac functional changes due to restriction / diastolic dysfunction / fibrosis

e) Arterial changes due to loss of vascular compliance

Page 178: Training Manual for Hemoglobinopathies and Hemophilia:

178

Pathophysiology

Iron accumulation in the heart leads to cardiac dysfunction. Heart is exposed to high levels of

non-transferrin bound iron which results in cardiac damage. Labile iron is quickly bound to

ferritin and degraded to hemosiderin which gets deposited in the myocites. Accumulation of

labile iron in myocytes results in damage to the membrane, cardiac failure, arrhythmias and

fibrosis. Cardiac dysfunction occurs predominantly due to iron overload but development of

hypothyroidism, hypoparathyroidism, hypogonadism can exacerbate cardiac dysfunctions.

Similarly deficiency of thiamine, vitamin D & selenium can worsen cardiac dysfunctions.

MRI cardiac T2* can quantitate cardiac iron stores which allows the clinician to modify

theiron chelation before the development of any cardiac symptoms. Iron chelator should be

selected depending on site of excess iron overload. Consult with a level 3 or 4 center.

Clinical Symptoms

Early symptoms include breathiness on exertion, dyspnea, edema. Clinician needs to examine

the patient for signs of left heart failure such as rales, orthopnea etc. Children with right heart

failure may develop distended neck veins, peripheral oedema, hepatomegaly etc. Heart failure

due to iron overload recovers with intensive chelation therapy.

Symptoms of palpitation may lead to arrhythmia which may be due to myocardial iron

overload or myocardial dysfunction or myocardial fibrosis. These patients need to be

evaluated by ambulatory or exercise electro cardiogram (ECG). Echocardiography (ECHO) is

required to assess various dimensions of heart chambers along with their function, doppler

flow assessment and morphology. MRI T2* indicates the iron overload in the heart with

cardiac complication. These patients need to be evaluated at level 2 / 3centers where the

cardiologist & above facilities are available. Initial management need to be carried at these

centers. Once these patients are stabilised & subsequent management can be carried out at

DCC as per advice of higher centers.

Pulmonary Hypertension

It develops due to interaction of multiple mechanical & biochemical factors leading to

smooth muscle proliferation in pulmonary vasculature (Fig. 6)

Diagnosis & Management

Echocardiography, complete pulmonary functions, higher resolution CT & CT angiogram are

essential for complete evaluation. All these tests will be available at level 2 or level 3. Initial

evaluation & management need to be undertaken at these centers. Subsequent management

can be undertaken at the integrated center for hemoglobinopathies center in consultation with

higher centers.

Page 179: Training Manual for Hemoglobinopathies and Hemophilia:

179

Arrhythmias

Children may present with palpitation or may be asymptomatic. Arrhythmias are life

threatening in presence of heart failure. Presence of Ectopic may trigger more sustained

arrhythmias. The evaluation of these children and management should be undertaken at level

2 or level 3. The subsequent management can be carried at DCC after they have been

stabilised at higher centers.

Management of cardiac complications

Prevention of cardiac toxicity

• Combination therapy with deferiprone (75-100 mg/kg) and deferoxamine (40-50

mg/kg/day sc over 10 – 12 hours thrice a week) is an option to reduce cardiac iron

overload and stabilize cardiac function. Combination therapy needs to be monitored

closely for toxicity and needs to be continued till the cardiac functions improve. Should

be done in collaboration with level 3 or 4 center.

In cardiac failure

• Ensure monitoring for arrhythmias and start on continuous deferoxamine therapy (50

mg/kg/day )as long as the patient has adequate urine output.

• Deferiprone 75 mg/kg/day, should be added in divided doses (TID), as soon as the patient

stabilizes and starts taking orally.

• Management includes diuretics, pressors, and antiarrhythmic drugs in consultation with a

cardiologist.

Key recommendation

• Thalassemia major patients with heart failure should be managed at (or in close

consultation with) a tertiary center experienced in thalassemia.

• Screen and treat endocrine and metabolic co-morbidities in thalassamia major patients

with ventricular dysfunction.

• Combination of two chelation therapy is helpful.

• Routine cardiac T2* MRI assessment represents the best available tool to prevent cardiac

dysfunction

• Echocardiographic screening for pulmonary hypertension should be performed

annually.

• Lifestyle choices that promote vascular health (absence of smoking, regular physical

activity, weight control, vegetable and nitrate rich diet) should be vigorously promoted in

thalassemia patients.

(D) Hepatic dysfunction Liver represents a major target of iron overload in atransfusion dependent thalassemia patient

and majority of patients with non-transfusion dependant thalassemia (NTDT). It is essential

Page 180: Training Manual for Hemoglobinopathies and Hemophilia:

180

to remove excess of hepatic iron as early as possible to protect liver and other organs like

heart & endocrine glands. Hepatitis viruses, especially hepatitis C virus (HCV) and hepatitis

B virus (HBV), should not be underestimated, as these patients are more prone to catch these.

The aggravating role of hepatotoxic co-factors, such as dysmetabolism and alcohol, should

also be kept in mind. We should avoid development of chronic liver disease, as that is an

obvious risk for development of cirrhosis and hepatocellular carcinoma.

Recommendations for prevention of hepatic dysfunction

Hepatic iron excess should be evaluated using a non-invasive strategy based on combined

serum ferritin values and T2* MRI data.

Liver biopsy is often not necessary.

Deferasirox is effective in producing a negative iron balance and decreasing hepatic

damage.

Combination therapy may be used if mono therapy cannot meet its objectives.

Incidence of hepatitis B has gone down significantly by institution of hepatitis B

vaccination at diagnosis, while hepatitis C can be picked up by screening these children

regularly every year.

Diagnosis and treatment of HCV and/or HBV chronic hepatitis should be done in

consultation with hepatologist at level 2 or level 3.

E) Endocrine complications 1. Growth Retardation

Growth retardation occurs almost invariably in TM subjects.

Significant size retardation is observed in stature, sitting height, weight, and biacromial

(shoulder) and bicristal (iliac crest) breadths.

After the age of 4 years, the longitudinal growth patterns display rates of growth

consistently behind those of normal controls. The bone age is frequently delayed after the

age of 6–7 years.

Growth retardation becomes severe with the failure of the pubertal growth spurt.

Key contributing factors to stunted growth is chronic anaemia, transfusional iron overload

and chelation toxicity.

Other important contributing factors include nutritional deficiencies (protein-calorie

malnutrition, vitamin D and A, zinc and carnitine deficiencies), growth hormone

deficiency (GHD) /insufficiency (GHI), insulin like growth factor-I (IGF-I) deficiency,

chronic liver disease, hypogonadism, hypothyroidism and psychosocial stress.

Diagnosis and investigations

Diagnosis requires careful clinical evaluation to establish:

Short stature –

o height below the 3rd centile for sex and age (based on national growth

charts) , and/or

o Slow growth rates- growth velocity expressed in cm/year, below 1SD for

age and sex (based on growth velocity charts), and /or

Page 181: Training Manual for Hemoglobinopathies and Hemophilia:

181

o Signs of other pituitary hormone deficiencies (e.g., gonadotrophins, GHD,

TSH deficiency).

Signs of other possible causes of retarded growth (nutritional deficiencies, chronic hepatic

disease, chronic heart failure).

Routine investigations include: Biochemical analysis, thyroid function (TSH and FT4),

bone age (X-ray of wrist and hand) and bone mineral density (BMD)

Recommendation-

Regular (six-monthly intervals) and accurate measurement of standing and sitting

height, pubertal staging (Table 2) and bone age

Prevention and treatment of growth abnormalities in patients with TM should

include:

Proper blood transfusion to maintain pretransfusion haemoglobin level between 9.5 to 10

g/dl.

Proper chelation to attain serum ferritin < 1000 ng/ml and to maintain this level.

Use of iron-chelators (Deferasirox) with lower toxicity on the skeleton and with better

compliance. Combination therapy may be used to control the serum ferritin if mono therapy

is unsuccessful.

Correction of nutritional deficiencies (protein-calorie, folate, vitamin D, vitamin A, zinc,

carnitine) when suspected.

Oral zinc sulphate supplementation should be given to patients with proven zinc deficiency.

Correction of hypersplenism.

Refer to endocrinologist in level 2 or level 3centers for proper diagnosis and management

of growth delay and GH treatment in patients with GHD.

2. DELAYED PUBERTY AND HYPOGONADISM

Delayed puberty and hypogonadism are the most obvious clinical consequences of iron

overload.

Delayed puberty is defined as the complete lack of pubertal development in girls by the age

of 13 and in boys by the age of 14 year. (see table 15)

Hypogonadism is defined in boys as the absence of testicular enlargement (less than 4 ml)

and in girls as the absence of breast development by the age of 16 years.

All efforts should be made to detect early and refer the case to level 2 or level 3 for

diagnosis & management.

Routine investigations include: biochemical analysis, thyroid function (TSH and FT4),

bone age (X-ray of wrist and hand) and bone mineral density (BMD).

Page 182: Training Manual for Hemoglobinopathies and Hemophilia:

182

Proper and timely management of pubertal delay in boys and girls with TM and appropriate

induction of puberty to attain normal pubertal growth spurt and normal bone accretion.

Tanner staging should be done annually after the age of 10 years.

Treatment can be carried at DCC in consultation with an Endocrinologist who are available

at level 2 or level 3.

3.HYPOTHYROIDISM

Excessive iron gets deposited in the thyroid gland leading to thyroid dysfunctions

Presents with stunted growth, delayed puberty, cardiac failure and pericardial effusion.

Free T4 and TSH are the key investigations

Sub-clinical hypothyroidism is a combination of high TSH with normal FT4 levels.

Overt hypothyroidism is a combination of high TSH with low FT4.

Investigation should be performed annually, beginning at the age of 9 years (unless

symptomatic hypothyroidism is observed) at level 2. Blood samples for these tests may be

sent for early diagnoses.

Additional tests may include the following at level 3centers:

Thyroid autoantibodies: anti-thyroid peroxidase and antithyroglobulin autoantibodies.

Thyroid antibodies performed in selected cases and to exclude autoimmunity, but usually

negative.

Ultrasonography of thyroid gland - to evaluate structural thyroid abnormalities.

Bone age, in selected cases.

Biochemistry including lipid profile.

Serum ferritin.

ECG and Echocardiogram (especially in severe cases).

Hypothalamic-pituitary magnetic resonance imaging (MRI), in selected cases.

Treatment

Good compliance with chelation therapy may prevent or improve subclinical

hypothyroidism (basal TSH 5 to10 mUI/ml).

Patients with overt hypothyroidism should be treated with L-thyroxine which can be easily

given at DCC.

4. HYPOPARATHYROIDISM

Seen in second decade of life of transfusion dependent patients with thalassemia major

Majority of patients show a mild form of the disease accompanied by paraesthesia.

More severe cases may demonstrate tetany, seizures or cardiac failure.

These children may be sent to level 2 or 3 whichever is nearer to DCC.

Investigations

Investigations should begin from the age of 16 years

Serum Calcium, Serum Phosphate levels

Page 183: Training Manual for Hemoglobinopathies and Hemophilia:

183

In children with low serum calcium & higher phosphate level serum parathormone should

be measured

Management

Aim is to prevent acute and chronic complications of hypocalcemia.

Treatment includes:

Oral administration of Vitamin D or one of its analogues.(caution - High doses of Vitamin D

may lead to hypercalcemia) .

Calcitriol, 0.25-1.0 µg, twice daily, is usually sufficient.

In patients with persistently high serum phosphate levels, a phosphate binder (except

aluminium) may be considered.

Tetany and cardiac failure due to severe hypocalcaemia require intravenous administration of

calcium, under careful cardiac monitoring, followed by oral vitamin D.No special diet is

recommended, but few odification in diet can be tried e.g. Diet rich in calcium. - dairy

products, green leafy vegetables.

Diet low in phosphorus-rich items. - avoiding carbonated soft drinks, which contain

phosphorus in the form of phosphoric acid. Eggs and meats also tend to be high in

phosphorus.

Monitoring- At the start of the treatment, weekly blood tests are required. These are

followed by quarterly plasma and 24-hour urinary calcium and phosphate measurements

5.DIABETES MELLITUS (DM)

Impaired glucose tolerance (IGT) & DM accounts for nearly 30% of endocrine disorders.

Often it occurs in patients who have been poorly chelated. However this complication may

occur in children who are well chelated & transfused secondary to chronic anemia earlier,

zinc deficiency and increased collagen deposition. Development of IGT starts in early second

decade along with the development of puberty and DM follows. Development of DM can be

prevented if these children can be identified by oral glucose tolerance test (OGTT) and are

subjected to intensive chelation therapy and further avoidance of chronic hypoxia. Thus it is

essential to get the OGTT regularly from 10th

year of life onwards.

Diagnostic criteria for glucose tolerance test (OGTT):

Fasting glucose >126 mg/dl is diagnostic of diabetes mellitus.

OGTT serum glucose at 2 hours > 200 mg/dl is diagnostic of diabetes mellitus.

OGTT serum glucose at 2 hours > 140 < 200 mg/dl indicates glucose intolerance

Monitoring glycaemic control

Daily home capillary glucose monitoring.

Urine ketones if blood sugar is above 250 mg/dl.

Fructosamine estimation every month.

Page 184: Training Manual for Hemoglobinopathies and Hemophilia:

184

HbA1c is not a reliable indicator of glycaemic control

Assessment of renal function.

Urinary microalbumin and protein.

Evaluation for retinopathy of diabetes mellitus.

Management

Initially it should be at level 2center or higher. Subsequently it can be carried at integrated

center for hemoglobinopathies and haemophilia under the guidance of endocrinologist /

higher center. Measures which need to be followed include (a) strict diet control (b) regular

physical activity (c) intensive iron chelation with two drugs. Children with persistently

elevated blood sugar despite above measures need insulin therapy or oral anti diabetic drugs

under the guidance of an endocrinologist.

These children need to be counselled about the disease and dangers of hypoglycaemic attacks

as well as diabetic coma. These children should know the sign of hypoglycaemia as well as

hyperglycaemia. They can take appropriate measures at home & should reach an integrated

center for hemoglobinopathies and hemophiliaorlevel 2 or higher center immediately for

further management.

F). Osteoporosis

Definition

Osteoporosis is defined when bone mineral density (BMD) T-score is < -2.5 leading to higher

risk of fracture following trivial trauma or minor falls.

Osteopenia is defined when BMD score is between -1 and -2.5.

Normal BMD T score > -1.0

All bones are undergoing dynamic process of resorption and deposition which is controlled

by parathyroid hormone (PTH), vitamin D, growth hormone, steroids, calcitonin etc. In

thalassemia marrow expansion results in cortical thinning and fragility of bones which is

controlled by multiple genetic & acquired factors. Endocrine complications such as

hypothyroidism, hypoparathyroidism, DM hypogonadism result in osteopenia or

osteoporosis. Iron deposition impairs osteoid maturation leading to osteomalacia, osteopenia

& osteoporosis. Vitamin C deficiency in TM increases the risk of osteoporotic fractures.

Reduced physical activity in TM predisposes to bone loss & subsequent osteoporosis.

Symptoms

Bone pains, weakness are early symptoms. Development of fractures on trivial trauma or

minor falls suggests development of osteoporosis.

Page 185: Training Manual for Hemoglobinopathies and Hemophilia:

185

Prevention & Treatment of Early Bone Loss

Physical activity in these children should be encouraged

Smoking to be avoided

Adequate calcium replacement (High intake of milk & milk products)

Vitamin D supplementation from early age

Adequate iron chelation from the beginning

Prevent the development of endocrinopathies

Annual BMD test by Dexa scan from 10 years of age. It can be performed at hip,

lumbar spine & distal radius. Development of osteopenia can be detected early by

Dexa scan.

Treatment

High index of suspicion& early detection on routine BMD are the only methods for early

diagnosis. These children need to be referred to level 2 / level 3centers for diagnosis &

management. It can be carried out at DCC with guidance from these centers. The

treatment for osteoporosis includes

Calcium & vitamin D supplementation

Use of Bisphosphonate

Calcitonim administration.

Table 15:Pubertal assessment according to Tanner

Penile Development Breast Development Growth of Pubic Hair

P1: Pre-pubertal B1: Pre-pubertal PH1: Pre-pubertal

P2: Early puberty

Enlargement of scrotum and

testes, 4-5 ml, little or no

enlargement of penis

B2: Early puberty

Breast bud stage

PH2: Early puberty

Sparse hair growth

P3: Mid-puberty

Enlargement of penis and

further growth of testes, 8-12

ml, and scrotum

B3: Mid-puberty

Breast and areolar

enlargement

PH3: Mid-puberty

Hair extends over the public

junction

P4: Advanced puberty

Enlargement of penis in length

& breadth. Increased

pigmentation of scrotal skin and

enlargement of testicles, 14-25

ml

B4: Advanced puberty

Areola and nipple project

separately from the contour of

the breast

PH4: Advanced puberty

Hair corresponds to adult

growth but less extensive

P5: Adult B5: Adult

Fully developed breast, the

PH5: Adult

Page 186: Training Manual for Hemoglobinopathies and Hemophilia:

186

areola no longer projects

separately from the breast

contour

Figure 5:Cardiac Complications

Iron Mediated Non Iron Mediated

Myocyte Failure

Endothelial Dysfunction

Arrhythmia

Pulmonary Hypertension

Vascular Stiffness

Thrombosis

Myocyte Scarring

Page 187: Training Manual for Hemoglobinopathies and Hemophilia:

187

Figure 6:Pathogenesis- Pulmonary Hypertension

References

1. Cohen AR, Glimm E &Porter JB. Effect of trans-functional iron intake on response to

chelation therapy in thalassemia major. Blood 2008;111: 583-87.

2. Thomsan AA, Genningham MJ, Singer SJ, et al. Red cell allo-immunization in a

diverse population of patients with thalassemia. Brit J Hematol. 2011; 153: 121-28

3. Pennell DJ, Udelson JE, Arai AE, et al. Cardiovascular function and treatment in

beta-thalassemia major: a consensus statement from the American Heart Association.

Circulation 2013; 128: 281-30.

4. Guidelines for the management of transfusion dependant thalassemia (TDT). Editors

Cappellini MD, Cohen A, Porter J et al. Thalassemia International Federation. 2014;

3rd

Edition.

+Vasoconstriction= Pulmonary hypertension

↑Cardiac output

Abnormalmechanical

forces

↑Hemolysis Liver dysfunction Splenectomy

↑ Platelet Aggregation

Endothelial dysfunction

↑Iron

↑LA Pressure

↓LV Dysfunction

Hypoxemia

Restrictive

lung disease

Pulmonary

Diffusion Block

Page 188: Training Manual for Hemoglobinopathies and Hemophilia:

188

Transfusion Reactions

Blood is a biologic product and great safety must be taken during the blood transfusion

process. Documentation of transfusion records are vital, time of starting, interrupting or

finishing of transfusion, vitals of patient, verification of bag and matching with records , all

of the steps must be recorded with date time and initials or signature. Any symptoms during

transfusion must be noted. The health care providers must be aware of other complications

which may occur later, and report these complications as they may require attention.

Transfusion reaction (TR) can be defined as an unintended response in a patient to

transfusion of blood components which prolongs hospitalization, is disabling or

incapacitating and increases morbidity or causes mortality. TRs can be broadly classified as

(i) acute or delayed (depending upon the time of occurrence) and (ii) immune or non-immune

(depending upon the pathophysiology).

Multi-transfused patients such as thalassemics are at a high risk of having transfusion

reactions due to the multiple and repeated transfusions they receive.

Page 189: Training Manual for Hemoglobinopathies and Hemophilia:

189

Vigilant supervision during transfusions is essential to identify transfusion reactions at the

earliest and manage them promptly so as to prevent any untoward effects.

The acute transfusion reactions will usually occur in the hospital setting, during or within a

few hours of transfusion and their clinical features are as follows:-

Table 16:

Common types of transfusion reactions.

SNo Type of Reaction Clinical Signa and symptoms

1 Acute Haemolytic Transfusion

Reaction(HTR)

Fever/chills, hypotension/tachycardia,

cola coloured urine, nausea, vomiting, pain in

flanks/back/abdomen/ chest

2 Bacterial Contamination Fever/chills, hypotension, nausea, vomiting,

dyspnoea, diarrhea.

3 Transfusion-related Acute Lung

Injury(TRALI)

Dyspnoea or cyanosis, fever, tachycardia,

hypotension

4 Febrile non hemolytic transfusion

reaction(FNHTR)

Fever, chills, rigors, cold, headache, nausea,

vomiting

5 Allergic/Anaphylactic reaction Pruritis, urticaria, flushing, angioedema,

hoarseness, stridor, wheezing, chest tightness,

dyspnoea, cyanosis, anxiety, nausea, abdominal

cramps, and diarrhea

As we can see above the signs and symptoms of acute transfusion reactions often overlap

and diagnosis may not be possible without a complete workup. In unconscious patients

oozing of blood from phlebotomy/surgery site or sudden hypotension should raise the

suspicion of a TR

During blood/blood component transfusion closely monitor the patient for the signs and

symptoms of a transfusion reaction.

Page 190: Training Manual for Hemoglobinopathies and Hemophilia:

190

IN CASE A REACTION IS SUSPECTED-

Stop the transfusion immediately and keep the IV line open with normal saline.

Institute immediate resuscitative care as per the nature of the transfusion reaction

Send the following to the Blood Bank

Blood bag and transfusion set

Post-transfusion blood sample-3-4ml inanEDTA vial

Reaction form with details of the nature of the reaction

If a hemolytic/ septic reaction is suspected: Send following investigations in addition-

Microbiology

1. Blood Culture-from patient within 1 hour of the onset of reaction

Hematology

1. Complete hemogram(CBC)

2. Plasma hemoglobin

3. Urine hemoglobin

4. Coagulation parameters

Biochemistry

1. Bilirubin(conjugated/unconjugated)

2. Serum electrolytes

3. Urea

4. S.Creatinine

5. LDH

Page 191: Training Manual for Hemoglobinopathies and Hemophilia:

191

Management of transfusion reactions-

1. FNHTR–Tab Paracetamol to be given and if this reaction occurs repeatedly, pre-

medication of antipyretic may be given for further transfusions. A cautionary note

being that an anti-pyretic may mask the signs/symptoms of other more serious TRs,

therefore vigilance becomes all the more important.

2. Allergic Reactions–An anti-allergic is to be given and transfusion re-attempted with a

fresh unit of the blood component.

3. Acute HTR–Give injection Lasix. Respiratory support as required and

Hydrocortisone if required.

4. Bacterial Contamination-Supportive care as per symptoms and antibiotics

5. TACO-Transfuse at a slower rate, give injection Lasix if required

6. TRALI- Respiratory support with supplemental oxygen, in severe cases intubation

and mechanical ventilation. Vasopressor agents in case of sustained hypotension.

There is no role of diuretics as the underlying pathophysiology is microvascular

injury.

At the Blood Bank, the following are done-

All documents are checked for any clerical error

In case of mix up, this prevents a second wrong unit from being transfused

Visual Inspection of blood bag

Direct Antiglobulin Test (DAT)

Repeat Grouping from pre and post Transfusion(Tx) sample

Repeat Cross-Match with pre and post-transfusion sample

Indirect Coomb’s Test (IAT)

Antibody identification if required

These tests will assess if the transfused was compatible or not and identification of antibody

if present. The blood bank will rule out any incompatible transfusion and identify any

incriminating antibody in case of incompatibility, which will prevent further reactions in case

of an immune hemolytic reactions. In the case of bacterial contamination, the organism

identified in patient and blood bag will be identical.

Page 192: Training Manual for Hemoglobinopathies and Hemophilia:

192

Transfusion reactions are a diagnosis of exclusion and the temporal relation with transfusion

is the most important factor in identifying these reactions.

Table 17:

Delayed Transfusion Reactions

S. No. Reactions Sign/Symptoms

1 Delayed-hemolytic

transfusion reaction (DHTR)

6-24 hours of Transfusion. Fever, jaundice,

declining hemoglobin, renal failure Sickle crisis

in sickle cell disease (SCD) patients

2 Post Transfusion Purpura

(PTP)

Severe thrombocytopenia (Platelet

count<10,000/µl) 1-24 days after the transfusion.

Purpura, bleeding, rarely fever

3 Transfusion-Associated Graft

vs Host Disease (TAGvHD)

2-30 days after Transfusion.

Whole body erythroderma, desquamation,

diarrhea, pancytopenia due to bone marrow

failure

4 Iron overload

Discussed in a separate chapter

5 Air Embolism

Breathlessness, chest pain, joint pain, confusion

6 Alloimmunization

Cross-match incompatibility

7 Transfusion Transmitted

Infections (TTIs)

Sero-positive for the infection may be –HIV,

HBV, HCV, etc

The delayed transfusion reactions may occur when a transfusion recipient has left the

hospital/transfusion center and therefore The person receiving transfusion as well as the

primary caregivers should be informed regarding signs and symptoms of delayed transfusion

reactions, so that they are aware and bring the transfusion recipient back to the hospital/

transfusion center in case of any delayed transfusion reaction.

The management of delayed TRs is as follows-

DHTR-If severe, treat as for acute HTR otherwise monitor and check for

alloantibodies in both serum and on RBCs, identify the incriminating antibody and

select appropriate antigen negative units for future transfusions.

PTP- Provide platelet antigen negative units and give IVIG, Corticosteroids as per

requirement and do Plasmapheresis if refractory to other treatments.

Page 193: Training Manual for Hemoglobinopathies and Hemophilia:

193

TAGvHD-Supportive therapy. The condition is difficult to treat and has a very high

mortality. Prevention by using irradiated blood components in at-risk patients should

be strictly adhered to.

Air Embolism- Secure airway, breathing, and circulation (ABC), including

cardiopulmonary resuscitation (CPR) when necessary. Shift to ICU setting for further

management

Alloimmunization- Identify the incriminating antibody and ensure the patient

receives antigen negative units for all future transfusions, whether the antibody is

detected or not in future testing. Give the Transfusion recipient an antibody card

stating the identity of the antigen against which the patient has developed an antibody.

TTIs- treat as per infection treatment protocols after due counseling

The early identification and management of TRs is extremely important to prevent morbidity

and mortality in the transfusion recipients. All transfusion reactions should be reported to the

blood transfusion services whether it impacts the management of the TR or not.

Prevention of transfusion reactions

Every hospital should have a hemo-vigilance programme.

1. Always recheck information and if possible recheck blood group prior to

administration. Positive patient identification is a step which has the highest

potential to prevent Acute TRs as most fatal Transfusions occur due to wrong blood

in tube (WBIT).

2. Do not leave blood after sent from blood bank or storage center, transfuse as soon as

possible. Maintenance of blood cold chain is essential to maintain the quality of

blood components. If a breach in the cold chain is suspected the component should

not be used for transfusion.

3. Always have a doctor on call to manage reactions.

4. Visual inspection of blood components prior to issue/transfusion for bloating,

discoloration, floaters, hemolysis, and leakage is essential.

5. Febrile non haemolytic transfusion reaction (FNHTR) – Due to presence of

leucocytes in the blood product. Use Leuco-depleted blood, if a patient is having

recurrent FNHTR give Paracetamol tablet before blood transfusion.

Page 194: Training Manual for Hemoglobinopathies and Hemophilia:

194

6. Allergic reactions – Urticaria, angioedema & severe cases anaphylaxis due to

plasma proteins, if recurrent , give washed RBC

7. Acute haemolytic transfusion reaction – Stop blood, start IV normal saline, give

injection Furesemide (lasix). Give oxygen, Hydrocortisone if required. Send sample

for complete hemogram, Direct Coombs test, PT, APTT, LFT, Urea, Creatinine.

Notify blood bank. Send blood bag & a fresh patients sample to the blood bank.

8. Volume overload & cardiac failure – slow transfusion, give injection Lasix if

required

9. TRALI (transfusion related acute lung injury) –This is acute lung injury by anti

neutrophilic antibodies present in donor blood. It causes capillary leak & ARDS

leading to tachycardia, dyspnoea, hypoxia within 4-6 hours of transfusion. Fatal

condition. Give oxygen, shift to Intensive care unit (ICU).

10. Transfusion transmitted infections (TTI) Check HBsAg, HCV & HIV every 6

months. NAT tested blood preferable.

11. Universal leucodepletion of blood components ( Platelet and RBC products).

12. Do not use plasma from multiparous women or persons with a history of blood

component transfusion (use only RBCs from this category of donors).

13. Patients with chronic anemia are at risk of volume overload and should be

transfused slowly to prevent TACO, also volume reduced blood components should

be used where ever possible. Never transfuse whole blood to raise hemoglobin.

Packed RBCs (PRBCs) should be used.

14. Irradiation in patients at risk of TAGvHD should be a part of the transfusion

protocol

Page 195: Training Manual for Hemoglobinopathies and Hemophilia:

195

ii) Sickle cell Disease

Basic recognition of symtoms and signs of sickle cell disease at level 1

(PHC/CHC)

Outline

Diagnosis

When to suspect?

Test to be done.

Referral to higher centre if required

Sickle cell disease is a group of inherited red blood cell disorders

When to suspect?

Any child presenting with one of the below mentioned symptoms should be screened for

sickle cell disease.

Page 196: Training Manual for Hemoglobinopathies and Hemophilia:

196

What to Investigate?

Screening test

Complete blood count( CBC) - Anemia with microcytic hypochromic red cell

indices

Peripheral smear- sickle shaped RBCs

Sickling test

Solubility test

Diagnostic test (if screeningtestpositive)

Hb electrophoresis/ HPLC test

What to do before referral

Maintain hydration

Treat Fever and Pain –with Acetaminophen or Non-steroidal anti-inflammatory

drugs (NSAIDs)

Infections (fever) - start antibiotic e.g. Amoxicillin

Anemia

Painful and swollen hands and feet

Recurrent unexplained body pain in chest, abdomen ,joints and bones

Frequent fever/infections

Jaundice

Bone pain

Sudden vision problems

Delayed growth

Stroke/paralysis

Large painful spleen with fever/increasing pallor

Non healing leg ulcer near ankle

Priapism

Non healing Leg ulcers

Family history

Page 197: Training Manual for Hemoglobinopathies and Hemophilia:

197

Important guidelines for management of Sickle Cell Disease

Sickle cell disease(SCD) is a group of inherited single-gene autosomal recessive disorders

caused by the ‘sickle’ gene, which affects haemoglobin structure and leads to complications

like pain, anemia, increased risk of infections, stroke etc.

If not recognized and managed, patients may even have life threatening situations.They

require lifelong treatment and monitoring to ensure the best health possible for the individual.

A suspected patient with sickle cell disease (SCD) will present with the following

complaints:

Persistent pallor , pain, fever, lethargy, jaundice, difficulty in breathing.

Complete physical examination of such a patient is done in the formof :

Vitals including B.P,SpO2 saturation

Hydration status.

Grading of pallor, presence of icterus.

Signs of infection including toxic look.

Systemic examination including Respiratory examination, splenic size, complete

neurological examination, examination of involved joint in pain crisis.

Initial investigationsto be done in a suspected case of sickle cell disease are:

CBC, Peripheral smear, Reticulocyte count, Solubility test.

HPLC analysis confirms the diagnosis and pattern of sickle cell disease.

Page 198: Training Manual for Hemoglobinopathies and Hemophilia:

198

Below ONE year of age only HPLC analysis should be done to diagnose Sickle

cell disease. Solubility. test and Hb Electrophoresis are not useful below 1 year of

age.

If transfusion given then no test for diagnosis of sickle cell disease should be

performed till 3 months after the transfusion.

Fig 7.

Evaluation of child with suspected sickle cell disease

Sodium metabisulfite slide test or solubility test.

(Cannot differentiate between sickle cell trait and disease)

(Not to be used in first year of life as a high level of ‘HbF’ is present)

Confirmation is done by Hb electrophoresis, isoelectric focusing,

highperformance liquid chromatography (HPLC), capillary electrophoresis or

DNA analysis.

(Diagnostic tests for SCD should not be done within 3 months of receiving blood

transfusions.)

Routine Outpatient Care, General Management and Monitoring

Enroll all newly diagnosed patients with sickle cell disease as well as newborns ofSCD

screening programme. Register in Hemoglobinopathy registry, enrol in integrated center

for hemoglobinopathies and haemophilia.

Ensure each family receives counselling for the disease and it'scomplications.

Ensure each family undergoes genetic counselling.

Obtain patient’s baseline clinical disease status via documenting crisis rate, usual

duration of pain crisis, usual home remedy and medication which parents give to subside

pain, and precipitating factors like past h/o acute chest syndrome and requirement of

ICU care and ventilation for it, transfusion received, baseline Hb level, episodes of

jaundice, documentation of co- morbidities, ongoing treatment.

Document HPLC pattern, HbF level.

The clinical evaluation and laboratory investigations should be done during the visit of

the patient to the clinic in the following fashion :-Keep monitoring as per assessment

Page 199: Training Manual for Hemoglobinopathies and Hemophilia:

199

sheet for integrated hemoglobinopathies center, document infectious complications or

other problems in chart too.

Assessment sheet for sickle cell disease (SCD) patients

Physical examination

Age

Frequency

All systemic examination

0-24 months

Every 15 days to 3 months

>24 months Every three months

Laboratory tests

CBC /Platelet/reticulocyte 3-24 months Every 3 months

>24 months Every 3 months

(Monthly if on Hydroxyurea)

Renal function tests 12months Annually

(every 3 months if

onHydroxyurea)

Liver function Tests 12 months Annually

(every 3 months if on

Hydroxyurea)

Urine Analysis for micro-

albuminuria

>6 years Annually

Screening Tests

Transcranial Doppler 2-18 years Annually , if borderline

repeat every 4 months, if

abnormal repeat every 2-4

weeks . MRI if abnormal

Pulmonary function >6 years Annually based on clinical

course

Echocardiogram >12 years Annually or depending on

symptoms.

Age at assessment and frequency of referral clinic visits:-

Clinic Age Frequency

Dental Examination Three to five years Annually

Ophthalmology Five years Annually

Adolescent counselling Fourteenyears As needed

Page 200: Training Manual for Hemoglobinopathies and Hemophilia:

200

PENICILLIN PROPHYLAXIS :

Given only to children <5 years- 125 mg BD till 2 years , then 250mg BD after 2 years

till the age of 5 years.

Discontinue penicillin prophylaxis in children with HbSS at age 5 years unless they have

had splenectomy, then continue lifelong.

FOLIC ACID SUPPLEMENTATION:

o less than 1 year of age, 2.5 mg daily

o more than 1 year of age, 5 mg daily

CRITERIA FOR ADMISSION

Immunizations (In addition to routine vaccines)

Acute illness requiring immediate medical care, including emergencies that need to

be defined :-

a. Temperature >38 0C

b. Pain inadequately relieved by home measures

c. Significant respiratory symptoms( cough, shortness of breath, chest pain)

d. Abdominal pain, distention, acute enlargement of spleen.

e. Any neurological signs or symptoms if any

f. Significant increase in pallor, fatigue, lethargy

g. Significant vomiting and diarrhoea

Acute illness characterized by any one of the above signs or symptoms listed above

can prove rapidly life threatening.

Thus it is essential that SCD patients have unimpeded access to health care

providers at the community where appropriate measures can be taken.

Page 201: Training Manual for Hemoglobinopathies and Hemophilia:

201

Vaccine Age/ Frequency

Pneumoccocal Vaccine (conj)

Pneumococcal Vaccine(Polysaccharide)

2 years, then 2 years following first dose.

>2 years

H-influenza vaccine <1 year , 3 dosesat an interval of 1 month; <2

years, 2 doses at an interval of 1 year

>2 years, 1 dose

Typhoid Vaccine >2 years,1stdose;every year, single dose

Influenza vaccine Annually

Acute clinical complications of sickle cell disease and theirmanagement

Manifestations Treatment

Painful event Dactylitis, pain in the sternum/ribs, pain in the

longbones ,priapism

Hydration, analgesia

Monitor pain

medication response

Infection Bacteraemia/sepsis, meningitis, osteomyelitis,

pneumonia ,malaria

Antibiotics, surgery

Anemia Splenic sequestration, transient aplastic

crisis,transfusion reaction,

Blood transfusion/

blood bank assessment

Organ damage Stroke, acute chest syndrome, splenic infarction,

renal papillary necrosis, priapism

Hydration, blood

transfusion, specialist

consultation

Pain management:

Children experience pain due to vaso-occlusive crisis. It may be difficult to assess pain,

visual analogue scale etc. are useful and should be used.

Pain medicines are necessary, start with paracetamol oral or I.V. if child has vomiting.

Follow WHO pain ladder, but avoid pethidene in patients of SCD.

In severe cases, may need admission and even opioids for pain management.

Older patients should be assessed at a level 3 or 4 centre for pain needs and

appropriately managed.

Chronic clinical complications of sickle cell disease and management

Manifestations Treatment

Kidneys Hyposthenuria, glomerular hyperfiltration,

glomerulosclerosis, albuminuria, end stage

renal disease

ACE Inhibitors, hydroxyurea,

dialysis, renal transplant

Page 202: Training Manual for Hemoglobinopathies and Hemophilia:

202

Heart/Lungs Restrictive lung disease, elevated tricuspid

jet velocity, pulmonary hypertension,

restrictive cardiomyopathy

Bronchodilators,

hydroxycarbamide,

transfusion

Brain Ischemic stroke, haemorrhagic stroke,

silent infarction, neurological decline

Transfusion,

hydroxycarbamide

Liver Jaundice, pigmented gall stones Cholecystectomy

Spleen Infarction, hypersplenism Splenectomy

Bones/skin Avascular necrosis, leg ulcers Physical therapy, cord

decompression, wound care.

Surgery

Eyes Retinopathy Laser therapy

Penis Impotence, infertility Surgery (if needed)

PARENTAL EDUCATION

a) Inheritance pattern, genetic risk

b) Splenic palpation

c) Sensitize to requirement of routine follow up and monitoring

d) Attention to fever and grade of fever

e) Vaccination

f) Recognition of pain

g) Home remedy for pain

h) Need of chronic medication like hydroxyurea

i) When to seek medical help

References

1. Zakari Y. Aliyu, Ashaunta R. Tumblin, and Gregory J. Kato. Current therapy of sickle

cell diseaseHaematologica. 2006 January ; 91(1): 7–10.

2. Expert panel report 2014. Evidence based management of Sickle cell disease

https://www.nhlbi.nih.gov/sites/default/files/media/docs/sickle-cell-disease-

report%20020816_0.pdf ( accessed May 2019)

Page 203: Training Manual for Hemoglobinopathies and Hemophilia:

203

Hydroxyurea therapy- principles and guidelines

Introduction Sickle cell disease (SCD) manifestations have great diversity, even though it isamonogenic

disease. In India SCD patients havethemilder Arab Indian haplotype with high HbFlevels as

compared to Africans. There are many factors responsible for diversity in the phenotype of

SCD, like co inheritance of alpha thalassemia and level of HbF. The overall mortality of

SCD patients in central India is 3.6 per 100 person years. The per patient mortality is 21.8 %

by 1.5 – 5 yrs of age in Gujarat whereas in central India it is higher, 82 % by 3 -4 yrs

Hydroxyurea isa ribonucleotide reductase inhibitor and it prevents the formation of

deoxyncleotides. It causes S phase arrest of all replicating cells. This leads to

myelosuppression,then stress erythropoiesis causesincrease in HbF production. HbF

ameliorates sickling improving both acute and chronic manifestations of SCD.

Even though many drugs lead to increase in HbF, HU has emerged as the most common drug

because of its advantages such as oral therapy, modest toxicity, predictable laboratory

evidence of increase in Hb F and proven clinical efficacy in acute sickle cell crisis,

prevention of organ dysfunction and survival benefits.

Hydroxyurea received US- FDA approval for the treatment of adults with severe SCD in

1998. On December 21, 2017, US -FDA granted regular approval to hydroxyurea for children

from 2 yrs of age with SCD. All the patients of SCD should be treated with HU as it not only

improves symptoms but also survival of these patients.

Page 204: Training Manual for Hemoglobinopathies and Hemophilia:

204

Studies of HU in both children and adults has shown reduced frequency of painful episodes,

Acute Chest Syndrome (ACS) events, less requirements for RBC transfusions and

hospitalizations compared to placebo. On long term follow up studies, it has also shown

survival advantage.

Recommendations:

1. Creating awareness and educatingall patients with SCA and their family members about

benefits of hydroxyurea therapy.

2. All children more than 9 months of age and all adults should be treated with

hydroxyurea. All the patients should be started on HU,however because cytopeniais one

of the preventable side effects, HU should be given wherever laboratory investigations

such as CBC is available and monitoring of SCD patients is possible .

3. To discontinue HU in pregnant or breast-feeding females.

4. To have established prescribing and monitoring protocol for Hydroxyurea therapy in

SCD patients.

5. Consider HU in patients with Hb S- β +thalassemia or Hb SC disease who have

recurrent sickle cell related pain that interferes with daily activities or quality of life.

6. As of now there is no evidence on the role of HU for primary stroke prevention in

adults (1D).

Note:Inthose. situations where the patient who is on hydroxyurea therapy is not likely

to follow up or cannot be monitored, the patient should be given fixed low dose

hydroxyurea therapy (10 mg/kg) without escalation.

Baseline investigation

Complete physical examination.

Hb electrophoresis and quantitative HbF % if available .

CBC with differential and reticulocyte count.

Liver function tests (AST, ALT Total protein).

Albumin, Total bilirubin.

Hydroxyurea Treatment Recommendations – modified as per NHLB 2014 and

BCSH guidelines

Page 205: Training Manual for Hemoglobinopathies and Hemophilia:

205

Renal function (BUN, Cr).

Pregnancy test for post- menarchal females.

Both adult male and female in reproductive age group should be counselled regarding

the need for contraception when on HU treatment

If clinically indicated may perform -

Test the Serum B-12 and folate (to ensure HU-related macrocytosis does not mask

deficiency).

Serum iron, TIBC, ferritin (to ensure HU related macrocytosis does not mask Fe-

deficiency).

Dosing

Optimal dosing of Hydroxyurea is still a debate.

Maximal therapeutic dose (MTD) is a dose which leads to greatest benefit without

toxicities or side effects where there is acceptable degree of marrow suppression.

As per our Indian study to start the patients on 10 – 15mg/kg/ day of hydroxyurea as

single morning dose which can be increased if there is no response or patient is still

symptomatic or patient has stroke and cannot be treated with regular transfusion

regimen.

Starting dosage of Hydroxyurea

Adults: 15 mg/kg/day (round up to the nearest 500 mg)

In patients with chronic kidney disease: 5–10 mg/kg/day

Infants and children: 10 -15 mg/kg/day

Dose escalation by 5 mg/kg every 4–6 weeks only in definite indication with all the

facility of regular follow up, laboratory monitoring and treating physician should be

able to manage the side effects. For dose escalation the flow chart has shown how to

monitor and change the dose of HU.

Essential Monitoring :

CBC and reticulocyte count at least every 4 weeks with the aim of maintaining ANC

>2 x10 9/L, platelet count ≥80,000/uL.

If neutropenia or thrombocytopenia, hold HU and monitor CBC weekly.

When the counts recover, restart the treatment at 5mg/kg/day lower than the previous

dose on which patient had cytopenias.

Page 206: Training Manual for Hemoglobinopathies and Hemophilia:

206

Dose escalation

Hydroxyurea dose escalation to the maximal tolerated dose (MTD) –this dose escalation is

only required if benefit not seen on the present dose.

Dose is escalated as tolerated to reach target absolute neutrophil count (ANC) of 2.0 X 10 9 /L

which is obtained during well or steady visits. Increase the dose by 5mg/kg/d every 8 weeks

up to maximum of 35mg/kg/day. During dose escalation CBC should be monitored at least

every 4 weeks

Once the HU dose is stabilized

CBC, with differential, reticulocyte count and platelet count every 2 – 3months should be

monitored

ALT, AST, BUN, S. Cr every 6 months.

Weight, BP and toxicity focused physical exam each visit.

Inquiry regarding side-effects and contraception.

Importance of adherence to the treatmentand the patient should be counselled frequently

How to prepare HU solution for paediatric patients (now liquid preparation also

commercially available)

Using sterile aseptic techniques- gloves etc empty the contents of the capsule and mix

with room temperature sterile water to get a concentration of 200 mg/ml. and the

solutions should be vigorously stirred.

Then the solution should be filtered to remove insoluble particles and flavoured syrup

can be added to produce a final concentration of 100 mg/ml.

Such preparations have chemical stability over 6 to 9 months in observation period in

a study. The Hospital pharmacy should be able to prepare this under strict aseptic

precaution.

Response to Hydroxyurea

Clinical response with HU treatment will take 3 to 6 months. Hence6 month trial

on the MTD is necessary before considering for drug discontinuation due to

failure of treatment.

Monitor RBC, increase in MCV and HbF levels (only if possible) for evidence of

consistent or progressive laboratory response. Increase in MCV can also be used

as evidence of compliance.

Page 207: Training Manual for Hemoglobinopathies and Hemophilia:

207

Majority of the non-response is mainly due to non- compliance to therapy or

failure to escalate to MTD.

HU should be continued during hospitalization unless patient has toxicity like

febrile neutropenia or bleeding due to thrombocytopenia

Fig 8 Toxicities of Hydroxyurea in SCD patients

Table 18. Short term toxicities of hydroxyurea

Common >10 % Occasional 1-10 % Rare <1% Very rare<<1%

Dose dependant

Neutropenia

Reticulocytopenia

Leukopenia

Thrombocytopenia Diarrhoea Allergic reaction

Anemia Gastritis Leg ulcer

Thinning of hairs Pancytopenia Increase in creatinine

Nausea Rash Increase in ALT

Non dose dependant

Nail / skin

hyperpigmentation

Anorexia Vomiting

Page 208: Training Manual for Hemoglobinopathies and Hemophilia:

208

Long term toxicities:

Leg ulcer is not shown to be common in patients on hydroxyurea but if the SCD

patienthas leg ulcer, he/sheshould be monitored carefully on HU treatment.

Hydroxyurea is not shown to increase the chance of myelodysplastic syndrome or

acute leukemia or any malignancy even after long term use of 17 years.

Fertility – On hydroxyurea many patients and partners had normal delivery.

Hydroxyurea should be stopped 3 months. prior to conception in females and in male

SCD patients whose partner is carrying, there is no need to discontinue. But if the

male partner has problems in conception it should be stopped for 3 months as some of

the studies have shown that HU affects the sperm motility.

Both male and female patients should be counselled regarding risks and benefits of

stopping Hydroxyurea prior to conception or in pregnancy.

To summarise, Hydroxyurea should be the standard of care for SCD patients and should be

included in the essentialdrugslist. Liquid formulation of hydroxyurea will be beneficial for

pediatric dosing. Early treatment will alter the natural course of the disease leading to

decrease in sickle associate acute events as well as chronic organ damage along with better

survival among these patients.

References

1. Bender MA. Sickle Cell Disease. 2003 Sep 15 [Updated 2017 Aug 17]. In: Adam MP,

Ardinger HH, Pagon RA, et al., editors. GeneReviews® [Internet]. Seattle (WA):

University of Washington, Seattle; 1993-2019.

2. McGann, Patrick T, and Russell E Ware. “Hydroxyurea therapy for sickle cell

anemia.” Expert opinion on drug safety vol. 14, 11 (2015): 1749-58.

3. Segal JB, Strouse JJ, Beach MC, et al. Hydroxyurea for the treatment of sickle cell

disease. Evid Rep Technol Assess. 2008; 165:1–95.

4. Amrana Qureshi,1 Banu Kaya,2 ShivanPancham, on behalf of the British Society for

Haematology. Guidelines for the use of hydroxycarbamide in children and adults

with sickle cell disease. A British Society for Haematology Guideline. British Journal

of Haematology, 2018, 181, 460–475.

Page 209: Training Manual for Hemoglobinopathies and Hemophilia:

209

5. Matthew M. Heeney, Matthew R. Whorton, Thad A. Howard, et al Chemical and

Functional Analysis of Hydroxyurea. Oral Solutions 2004. J PediatrHematol Oncol

2004;26:179–184.

6. Jain, D.L., Sarathi, V., Desai, S., Bhatnagar, M. &Lodha, A. (2012) Low fixed dose

hydroxyurea in severely affected children with sickle cell disease. Hemoglobin, 36,

323–332.

7. Evidence-Based Management of Sickle Cell Disease, Expert Panel Report, 2014:

Guide to Recommendations. U.S department of health and human services.National

Institute of health. National heart lung and blood institute.

Page 210: Training Manual for Hemoglobinopathies and Hemophilia:

210

Common complications in Sickle cell disease and their management

Clinical features of SCD are markedly variable among individuals, depending on the form of

the disease (HbSS, or HbSβ-thal).

Goal of Management of SCD:

To improve quality of life and life expectancy of the affected individuals.

Objectives of Clinical Management of SCD:

a) Prevent and reduce the number of crises and complications

b) Treat crises and complications promptly and effectively

c) Promote a healthy lifestyle

Clinical Presentations

The major acute complications of SCD include the following:

A. Infections

B. Severe anemia (eg, due to splenic sequestration, aplastic crisis, or hyperhemolysis)

C. Vaso-occlusive phenomena

1. Acute vaso-occlusive pain

2. Stroke

3. Acute chest syndrome

4. Renal infarction or medication toxicity

5. Dactylitis or bone infarction

6. Myocardial infarction

7. Complications related to pregnancy

8. Priapism

Page 211: Training Manual for Hemoglobinopathies and Hemophilia:

211

9. Venous thromboembolism

Major chronic manifestations are:

1. Chronic Pain

2. Anemia

3. Neurologic deficits or seizure disorder

4. Pulmonary conditions

5. Renal impairment and hypertension

6. Osteoporosis and complications of bone infarction

7. Cardiomyopathy with diastolic dysfunction

8. Hepatotoxicity and pigmented gallstones

9. Chronic leg ulcers

10. Proliferative retinopathy

Sickle Cell Crisis

This refers to a worsening, over a short period of time, of the symptoms and signs of SCD;

usually associated with pain and/or anemia.

Comprehensive management of the patient in crisis has two major aspects namely:

a) Treatment of acute clinical problems during the episode of crisis; and

b) Arrangements to ensure continued care of the person in (post-crisis) healthy state.

Predisposing Factors for Sickle Cell Crisis:

a) Exposure to cold / drenched by rain

b) Physical exertion

c) Dehydration

d) Injury (including surgical injury)

e) Psychological stress

f) Idiosyncratic (peculiar to the individual)

g) Idiopathic (unidentified)

h) Infections/infestations

Initial Evaluation of a SCD Patient in Crisis should include:

1. History of pain, self-assessment of pain, and prior treatment taken before arrival

atthehospital

2. History of usually effective analgesics

3. History of drug allergies

Page 212: Training Manual for Hemoglobinopathies and Hemophilia:

212

4. Assessment of vital signs: blood pressure, heart rate, respiratory rate, oxygen

saturation (administer oxygen if O2 saturation<90%) and temperature.

5. History of increasing jaundice and passage of coke-coloured urine

6. Assessment of areas of bone tenderness.

Table 19:Initial Treatment of Patient in Sickle Cell Crisis

Pain relief (preferably in a comfortable and quiet environment)

Optimal hydration

Identification and treatment of infections and/or other cor-morbid conditions

Blood transfusion if necessary

Address specific clinical problems e.g. stroke, acute chest syndrome, priapism.

Table 20:Investigationsthat Influence Immediate Management of Sickle Cell Crisis

Urgent Hb/PCV, WBC**, Platelet and Reticulocyte counts, ESR

Examination of Blood Film especially for malaria parasites.

Plasma Bilirubin Level (total & conjugated)

Serum Urea, Electrolytes, Creatinine & C-Reactive Protein (CRP)

Infection Screening (blood, urine, stool, sputum etc ) as necessary

Abdominopelvic ultrasound scan as necessary

Chest X-Ray

Pulse Oximetry (Arterial Blood Gases if SaO2 <92%)

Transcranial Doppler Ultrasonography

CT / MRI

ECG / ECHO

Although a rise in the plasma level of acute phase reactants (e.g. C-reactive protein) may

indicate the transition from steady-state to crisis, the diagnosis of crisis is made on clinical

grounds. The frequency of crisis varies from person to person and from time to time in the

same person.

Acute pain syndrome

This is caused by obstruction of blood vessels, which leads to tissue ischemia /infarction and

pain, which is of sudden onset, with or without history of a predisposing factor, hence also

Page 213: Training Manual for Hemoglobinopathies and Hemophilia:

213

called a veno-occlusive crisis (VOC). The bones are most frequently involved, and they

become mildly to extremely painful and may also become swollen or tender, or both.

Table21:Management of acute pain crisis/ veno-occlusive crisis (VOC) :

Treat pain rapidly before full medical assessment. Fluid Management in Painful Crisis.

Avoid over hydration

Adults Children

1.5 L/m2/day OR 3L/day 100 – 120 ml/kg/day (can be given orally if there is

no vomiting or if patient can drink that volume

5% dextrose or dextrose saline is preferred. Avoid hypertonic fluid such as 10% dextrose.

Transfuse blood if Hb < 6g/dl, or > 2g/dl below steady-state value, or if symptomatic severe

anaemia.

If the patient has features of infection, send relevant samples for microbiology and start

broad-spectrum antibiotics, and anti-malarials in malaria endemic regions.

Determine characteristics, associated symptoms, location, and intensity of pain based on

patient self-report and observation. If the VOC pain is atypical, investigate other possible

etiologies of pain.

When oxygen saturation <95 percent on room air, administer oxygen.

In adults and children with VOC, use adjunctive non pharmacologic approaches to treat pain

such as local heat application and distraction.

Table 22:Treatment of Acute Pain in Sickle Cell Disease

Degree of Pain Opioid Naive Opioid Tolerant

Mild Adult:

Dihydrocodiene tablet 30 mg 4 hourly plus

Paracetamol 1g every4 hourly

Immediate-release oral

formulation of stronger

opiate e.g. morphine 4

hourly

Children:

Paracetamol 20mg/kg 4 to 6hourly

with/without Ibuprufen 10mg/kg 8hourly Or

diclofenac 1mg/kg 8hourly

Page 214: Training Manual for Hemoglobinopathies and Hemophilia:

214

Dihydrocodiene 1mg/kg 8hourly

Moderate/Severe Adult:

Diamorphine 2.5-5mg/4hourly s.c. Or

morphine 5-10mg stat or pentazocine 15 mg

8-hourly.

Inj diclofenac 75mg 1-2 hourly after opioids

Inj tramadol 100mg 1-2 hourly after

NSAIDs

Injection of opiate

diamorphine 10-

20mg/2-4 hourly s.c.

Injection of opiate e.g.

pentazocine 30 - 60 mg

8-hourly

Children:

Oral morphine 0.4mg/kg or diamorphine

0.1mg/kg in IV infusion or IM/SC stat toget

immediate pain relief.

Then maintenance: Slow release morphine

1mg/kg (rounded up to 5mg) every 12 hours

with oral morphine 0.3mg/kg every 3hours

as necessary. Or oral morphine 0.4mg/kg Or

pentazocine at appropriate doses

Inj diclofenac 1mg/kg after opoids

Inj tramadol 100mg 1-2 hours after NSAIDs

Avoid meperidine unless it is the only effective opioid for an individual patient.

Assess pain relief every 15-30 minutes if patient is awake and while he/she is left in a

comfortable place

Give anti-emetics if indicated: procholorperazine 250 μg/kg t.d.s. or cyclizine 12.5-25

mg t.d.s.

Monitor pain, sedation, vital signs, respiratory rate, oxygen saturations: every 30 min

until pain is controlled and patient is stable; then every 2 hours

Monitor opioid induced respiratory depression

Discharge patient when pain is improving or controlled with reduced dose of oral

analgesia e.g tramadol and NSAIDs

Arrange for home and outpatient follow-up appointment as applicable

Page 215: Training Manual for Hemoglobinopathies and Hemophilia:

215

Sequestration Crisis

This is pooling (sequestration) of a large proportion of blood in the spleen or liver. Usually

occurs in children < 6 years and some HbSβ-thal adults; unusual in HbSS adults.

Features of sequestration crisis include:

a) Marked pallor

b) Precipitous fall in Hb Level

c) Sudden, progressive enlargement of the spleen / increase in abdominal girth

d) Hypovolaemic shock due to reduced circulating volume

e) Reticulocytosis in blood film (differentiate sequestration from aplastic crisis)

Treatment of Sequestration Crisis:

In consultation with a sickle cell expert, transfuse patientswho have acute splenic

sequestration and severe anemia to raise the hemoglobin to a stable level, while

avoiding over-transfusion.

In consultation with a sickle cell expert, address the performance and timing of

splenectomy in people with recurrent acute splenic sequestration.

Teaching parents, school teachers and care givers how to palpate for an enlarged spleen

or liver

Prevention of Sequestration Crisis:

This is advised after 2 episodes, using one of 2 approaches:

Splenectomy after two episodes of sequestration crises occurring in less than six months

apart

Hypertransfusion in malaria endemic regions.

Hyperhemolytic crisis

Hyperhemolytic crisis refers to the sudden exacerbation of hemolysis with worsening anemia

despite ongoing reticulocyte production. This is a rare complication. The proposed

mechanisms are:

1. Occult splenic sequestration or aplastic crisis detected during a period of resolving

reticulocytosis.

2. Delayed hemolytic transfusion reaction ("bystander hemolysis")

Page 216: Training Manual for Hemoglobinopathies and Hemophilia:

216

3. Accelerated hemolysis in association with acute vaso-occlusive events

4. Infections and/or drug exposure (e.g. co-existence of G6PD defeciency)

Hyperhemolytic crisis is potentially fatal if the cause of hemolysis is not addressed quickly.

Corticosteroids (methylprednisolone 500 mg/day for two days) and intravenous

immunoglobulin (IVIG; eg, 0.4 g/kg daily for five days) are the mainstays of treatment.

Additional compatible transfusions should be limited to situations of life-threatening anemia.

Stroke

Patient with SCD who presents with a new neurologic defect (focal or nonfocal) or altered

level of consciousness is considered to be having potential stroke.

The incidence of stroke is three hundred times higher in HbSS than HbAA children.

Risk Factors for Stroke in SCD:

a. Age (Children of 2-16 years of age are more commonly affected)

b. Blood velocity >200cm/s in distal internal carotid, middle or anterior cerebral

artery

c. Previous Transient Ischemic Attack

d. Steady state Hb < 7 g/dl or Neutrophils > 10 x 109/l

e. Nocturnal hypoxemia + Sleep apnoea

f. HbSS sibling with CVA (reflecting genetic factors)

Differential Diagnoses of Stroke in SCD:

a) Acute ischemic stroke

b) Acute hemorrhagic stroke

c) Transient ischemic attack (TIA)

d) Cerebral venous sinus thrombosis

e) Acute meningitis

f) Seizure, particularly when associated with prolonged postictal paralysis (Todd's)

Prevention of Primary Stroke:

a) All children aged 2-16 years should have trans-cranial ultrasonography to identify

those with a high risk of developing stroke. Repeat in 3 months in cases with a velocity

of 170-199cm/s. If blood velocity ≥ 200 cm/s in the anterior or middle cerebral artery

Page 217: Training Manual for Hemoglobinopathies and Hemophilia:

217

detected on two occasions 6 wks apart is an indication for a transfusion programme to

prevent Primary Stroke.

b) Another indication for Primary Stroke prevention is occurrence of transient ischemic

attacks (TIAs).

c) In those identified with the high risk, stroke should be prevented with hydroxyurea or

transfusion

d) If blood transfusion is used, to be effective, the circulating HbS should be reduced to

<30%

Features of Stroke in SCD:

e) Headache

f) Vomiting

g) Seizures, various sensory or motor neurological deficits: hemiplegia/paresis,

paraplegia/paresis, monoplegia /hemiparesis, loss of hearing, etc.

h) Rarely, sudden reduction or loss of consciousness, or sudden death

i) Magnetic Resonance Imaging (preferred over CT scan) may show acute infarct or

bleed in the brain.

Management of Acute Stroke in SCD:

a) Initial Assessment and Care:

Brief history and physical examination; including the nervous system to distinguish

between symptoms due to pain from those due to weakness (paresis or paraplegia)

Stabilization, support, and monitoring of vital signs as necessary; maintain normal

temperature

Good oxygenation

Careful hydration at two-third of the calculated maintenance volume in case the

Syndrome of Inappropriate ADH Secretion (SIADH) develops

Hydroxyurea can be used if hypertransfusion is not feasible.

b) Laboratory Evaluation (Investigations):

Full blood count with reticulocyte counts,grouping and extended cross-matching,

Blood electrolytes, creatinine, urea, tests for meningitis if suspected clinically.

c) Neuroimaging: MRI with MRV/MRA. DW MRI is highly sensitive in detecting early

ischemic changes.

Page 218: Training Manual for Hemoglobinopathies and Hemophilia:

218

d) Red Cell Transfusion:

Initially, simple (top-up) or exchange transfusion; to be followed by a regular

transfusion programme, preferably exchange, to keep the proportion of HbS below

30% in the patient’s blood; indefinitely, to prevent Secondary Stroke.

e) Iron Chelation: Start when the serum ferritin concentration is 1000 ug/L or greater.

f) Non-medical Interventions and Rehabilitation:

To improve quality of life of People Living With Sickle Cell Disease (PLWSCD), the

use of devices that facilitate mobility, education and self-help skills should be put in

place.

Priapism

This refers to a prolonged painful penile erection, not associated with sexual stimulation.

Types of Priapism:

a) Stuttering, lasts < 4 hr; resolves spontaneously

b) Major or fulminant, with duration of > 4 hrs ; less frequent.

Onset: Mostly in the early hours of the morning and at nights.

Complications:

a) Erectile dysfunction

b) Psychosocial problems.

During clinical assessment, ascertain if there is urinary retention and whether the glans penis

is soft or turgid. This influences treatment. If the patient is able to pass urine and the glans is

soft, then the corpus spongiosum is probably NOT affected, and a glans-cavernosa shunt may

confer clinical benefit.Otherwise, a surgical shunt between the dorsal vein of the penis and

the corpora cavernosa may be more beneficial.

Management of Stuttering (Minor) Priapism:

a) Give slow release tablets of etilefrine. The effect lasts for 8-9 hrs. Start with 25mg

daily taken by 10-11 pm. If clinical response is unsatisfactory after 2 weeks, increase

the dose to 50mg daily. If response still not satisfactory, increase the dose by 25 mg

every 2weeksup to a maximum dose of 100mg .

Page 219: Training Manual for Hemoglobinopathies and Hemophilia:

219

b) If the total daily dose of etilefrine is greater than 50mg, it is recommended that 50mg

be taken by 10-11 pm, and the rest by 4-5 pm.

c) Monitor BP and erectile function in people on etilefrine, because it is a vaso-

constrictor.

a) If etilefrine is not effective in preventing priapism, cyproterone(anti-androgen) is

added.

Management of Major Priapism in Sickle Cell Disease:

a) Give pain killers to comfort the patient

b) Give fluids (oral or intravenous)

c) Urosurgeons should aspirate and irrigate corpora cavernosa with 6-10 mg of

injectable etilefrine diluted in 20mls of saline. If there is no response (detumescence)

after 1 hour, repeat irrigation with etilefrine. Phenylephrine may be used in place of

etilefrine, but is less effective.

d) If repeat irrigation of cavernosa with etilefrine does not lead to detumescence after

1hour, the uro-surgeons should create shunts between the corpus spongiosum and

corpora cavernosa to drain deoxygenated and sickled blood sequestered in the

cavernosa into the spongiosum, and from there to the general circulation.

b) Exchange blood transfusion is usually not effective for established episodes of

fulminant priapis

Management of Chronic Leg Ulceration in Sickle Cell Disease

Chronic leg ulceration occurs in less than 10% of people with SCD, usually in patients who

are older than 12 years.

Clinical Features:

a) Typical ulcerations are situated around the medial or lateral malleolus of one or both

ankles

b) It arises spontaneously, although a history of preceding trauma may occasionally be

obtained

c) Healing is very slow and recurrence or breakdown of healed ulcers is very common.

Treatment:

a) Daily wound dressing

b) Rest and elevate the affected limb

c) Treat any associated infection

Page 220: Training Manual for Hemoglobinopathies and Hemophilia:

220

d) Autologous skin graft.

Factors that Promote Healing Leg Ulcers:

a) A clean wound

b) Daily dressings to keep the surface fresh and clean,

c) Rest.

Factors that reduce Healing of Leg Ulcers:

a) Walking and running long distances

b) Infection of ulcer surface

c) Scabs on the ulcer surface

d) Underlying osteomyelitis.

Infections

Globally, infection is the leading cause of death among SCD patients in developing countries.

It is critical that fever alone is taken seriously in these individuals and considered a potential

emergency situation. Fever associated with pain should not be considered a VOC until

infection is ruled out.

Predisposing factors:Functionalasplenia (hyposplenism), impaired complement activity,

zinc deficiency, iron overload, hypoventilation and the presence of necrotic tissue.

In addition, infection is a common precipitant of an acute crisis, and prompt treatment with

appropriate antibiotics is therefore essential.

Persons with SCD have normal T cell and B cell function, so the risk of acute infection is

generally limited to capsulated micro-organisms, salmonella, or other enteric pathogens.

Opportunistic infections are infrequent.

Viral infections may also be more virulent in individuals with SCD (eg, parvovirus, H1N1

influenza, Zika virus), possibly due to increased sickling and an enhanced inflammatory

response.

Management

1. In people with SCD and a temperature ≥101.3°F (38.5°C), immediately evaluate with

history and physical examination, complete blood count (CBC) with differential,

reticulocyte count, blood culture, and urine culture.

Page 221: Training Manual for Hemoglobinopathies and Hemophilia:

221

2. Promptly administer empiric parenteral antibiotics that provide coverage against

Streptococcus pneumoniae and gram-negative enteric organisms. Subsequent outpatient

management using an oral antibiotic is feasible in people who do not appear ill.

3. Hospitalize people with SCD and a temperature ≥103.1 °F (39.5 °C) and who appear ill

for close observation and intravenous antibiotic therapy.

4. In people with SCD whose febrile illness is accompanied by shortness of breath,

tachypnea, cough, and/or rales, manage according to the preceding recommendations

and obtain an immediate chest x ray to investigate for ACS.

5. In febrile people with SCD who have localized or multifocal bone tenderness, especially

when accompanied by erythema and swelling include bacterial osteomyelitis in the

differential diagnosis and manage accordingly.

6. Worldwide, malaria is a common cause of morbidity and mortality in children with

SCD.

Antibiotic prophylaxis

Penicillin VK 125 mg twice daily orally for children under 3 years of age and 250 mg bid for

children older than 3 years of age is recommended, and continued until adolescence.

Erythromycin is recommended for patients who are allergic to penicillin.

Immunisation

a) Pneumococcal vaccine. Adults and children >2 years of age should receive the

unconjugated vaccine (Pneumovax 23). Conjugated vaccine (Prevenar 13) is provided

to children under the age of 2 years as part of the national immunisation schedule.

Children who have missed the conjugated vaccine should have two doses of the

conjugated vaccine followed by a dose of the unconjugated vaccine, at 6 - 8-week

intervals.

b) A single dose conjugated meningococcal C vaccine should be given.

c) Haemophilus influenzae type B vaccine. If not given as part of the childhood

immunisation programme, a single dose should be given.

d) Annual influenza vaccination is recommended.

Hypersplenism

Hypersplenism classically refers to splenomegaly and any combination of anaemia,

leucopenia and/or thrombocytopenia, with compensatory bone marrow hyperplasia for a

sustained period, and improvement after splenectomy.

Page 222: Training Manual for Hemoglobinopathies and Hemophilia:

222

Because of pre-existing anemia; thrombocytopenia and ‘abnormally normal’ leucocyte count

are the main haematological signs. Transfusion efficiency is usually reduced in SCA patients

with hypersplenism.

Management of splenomegaly and hypersplenism:

Isolated mild splenomegaly warrants no specific management apart from parental education

on the risk of acute splenic sequestration.

When the spleen is markedly enlarged with biological signs of hypersplenism, along with

frequent life-threatening episodes of sequestration crisis, poor growth, bone marrow

hyperplasia and abdominal distension, a supportive transfusion programme can be initiated or

splenectomy performed if age allows. Violent sports should be restricted in order to avoid

traumatic splenic rupture.

Splenectomy in SCD

Splenectomy needs to be considered at an individual level, balancing relative risks and

benefits by taking into account the indication, the medical environment, parental wishes and

reliability, and the risks related to alternative treatment, such as chronic transfusions. In most

patients with SCD it is not required.

Laparoscopic splenectomy has become the procedure of choice for most children requiring

splenectomy. Partial splenectomy has also been proposed as an alternative treatment in very

young children. Following splenectomy, leucocytosis and thrombocytosis are common

findings and generally return to expected basal levels within a year.

Acute Chest Syndrome

ACS is the leading cause of death in SCD, and is characterised by:

Fever – temperature >38.5oC

Respiratory symptoms and signs, including cough, chest pain, dyspnoea or

wheezing

A new pulmonary infiltrate on chest X-ray (e.g. consolidation, segmental

changes, etc.)

ACS may begin as a pain crisis affecting the ribs, chest or shoulders.

Page 223: Training Manual for Hemoglobinopathies and Hemophilia:

223

Complications of an ACS:Respiratoryfailure, central nervous system injury (from anoxia,

infarction or hemorrhage), seizures, non-cardiogenic pulmonary oedema and multiorgan

failure (from hypoxia, resulting in widespread sickling and VOC). Patients with ACS are best

cared for in a high-care facility.

Infection is a more common cause of ACS in children compared with adults; infarction and

bone marrow fat embolism are more common in adults relative to children. Repeated

episodes of ACS are associated with chronic sickle lung disease.

To reduce the risk of mortality, especially in adults, all the contributing factors for ACS are

assumed to be present and treatment is given accordingly.

Table 23:Clinical Features of Acute Chest Syndrome

Symptoms Signs

Fever Breathlessness

Cough Wheezing

Chest pain Nasal flaring

Hemoptysis (though rare) Tachypnea

Hypoxia

Tachycardia

Normal chest findings

Dullness to chest percussion

Crepitations in lung

Initial chest x-rays may not show new opacities in the lungs, serial radiographs are required

in such situations. The enzyme secretory Phospholipase A2 (sPLA2) is increased before the

clinical features of ACS become obvious, and is a laboratory predictor of this condition.

Plasma level of C-reactive protein (CRP) correlates very well with that of secretory

phospholipase A2 and in ACS serum CRP is usually >5mg/L

It is important to distinguish between Acute Chest Syndromes from vaso-occlusive crisis

involving the bones of the chest for which the treatment is different.

Treatment:

1. Admit to hospital

Page 224: Training Manual for Hemoglobinopathies and Hemophilia:

224

2. Oxygen is beneficial if there is hypoxemia; may not if blood oxygen saturation is

normal

3. Exchange Blood transfusion (EBT), but if not possible, do a top-up so as to facilitate

oxygen delivery to the tissues

4. Broad-spectrum antimicrobials, including clarithromycin for mycoplasma and

Chlamydia

5. Bronchodilators, such as nebulized salbutamol to improve oxygen delivery to the

Lungs

6. Anticoagulant therapy, especially if marrow fat or thromboembolism is suspected

7. Analgesics, to reduce chest pain which may inhibit breathing and impair oxygenation

8. Cautiously maintain optimal hydration (to prevent lung edema).

Prevention of ACS:

Hydroxyurea therapy or hypertransfusion should be commenced after recovery to prevent

recurrence.

Evidence-Based Recommendations for Use of Hydroxyurea Therapy

1. In adults with sickle cell who have ≥3 moderate to severe pain crises associated with

sickle cell disease (SCD) during a 12-month period, initiate treatment with

hydroxyurea.

2. In adults with SCD who have sickle cell–associated pain that interferes with daily

activities and quality of life, initiate treatment with hydroxyurea.

3. In adults with SCD who have a history of severe or recurrent acute chest syndrome

(ACS), initiate treatment with hydroxyurea.

4. In adults with SCD who have severe symptomatic chronic anemia that interferes with

daily activities or quality of life, initiate treatment with hydroxyurea.

5. In infants 9 monthsof age or older, in children, and in adolescents with SCD, offer

treatment with hydroxyurea regardless of clinical severity to reduce complications

(e.g, pain, dactylitis, ACS, anemia) related to SCD.

6. In adults and children with SCD who have chronic kidney disease and are taking

erythropoietin, add hydroxyurea therapy to improve anemia.

7. Discontinue hydroxyurea therapy in women who are pregnant or breastfeeding.

8. Use an established prescribing and monitoring protocol to ensure proper use of

hydroxyurea and maximize benefits and safety.

Page 225: Training Manual for Hemoglobinopathies and Hemophilia:

225

9. In persons with HbSβ+-thalassemia diseasewho have recurrent SCD-associated pain

that interferes with daily activities or quality of life, consult an SCD expert for

consideration of hydroxyurea therapy

10. In persons not demonstrating a clinical response to appropriate doses and duration of

hydroxyurea therapy, consult an SCD expert

Recommended Laboratory Tests before Starting Therapy

Complete blood count (CBC) with white blood cell (WBC) differential, reticulocyte

count, platelet countred blood cell count(RBC) and mean corpuscular volume (MCV)

Quantitative measurement of fetalhemoglobin if available (eg, hemoglobin

electrophoresis, high-performance liquid chromatography)

Renal and liver function tests

Pregnancy test for women

Initiating and Monitoring of Hydroxyurea Therapy

1. Baseline elevation of fetalhemoglobin should not affect the decision to initiate

hydroxyurea therapy.

2. Both males and females of reproductive age should be counseled regarding the need

for contraception while taking hydroxyurea.

3. Starting dosage for adults (500mg capsules): 15mg/kg/day (round up to the nearest

500mg); 5-10mg/kg/day if patient has chronic kidney disease.

4. Starting dosage for infants and children: 20mg/kg/day.

5. Monitor CBC count with WBC differential and reticulocyte count at least every 4

weeks when adjusting dosage.

6. Aim for a target absolute neutrophil count more or equal to 2000/μL; however,

younger persons with lower baseline counts may safely tolerate absolute neutrophil

counts down to 1250/μL.

7. Maintain platelet count more than or equal to 80 000/μL.

8. If neutropenia or thrombocytopenia occurs:

Temporarily stop hydroxyurea dosing.

Monitor CBC count with WBC differential weekly.

When blood counts have recovered, reinstitute hydroxyurea at a dose of 5

mg/kg/day lower than the dose given before onset of cytopenia

Page 226: Training Manual for Hemoglobinopathies and Hemophilia:

226

9. If dose escalation is warranted based on clinical and laboratory findings, proceed as

follows:

Increase by 5-mg/kg/day increments every 8weeks.

Give until mild myelosuppression (absolute neutrophil count of 2000-

4000/μL) is achieved, up to a maximum of 35mg/kg/day.

Once a stable dose is established, laboratory safety monitoring should include

CBC count with WBC differential, reticulocyte count, and platelet count every

2-3 months.

10. People should be reminded that the effectiveness of hydroxyurea depends on their

adherence to daily dosing; they should be counseled not to double up doses if a dose

is missed

11. A clinical response to treatment with hydroxyurea may take 3-6 months; therefore, a

6-month trial on the maximum tolerated dose is required prior to considering

discontinuation due to treatment failure (whether due to lack of adherence or failure to

respond to therapy).

Monitor RBC MCV and fetalhemoglobin levels for evidence of consistent or

progressive laboratory response.

12. A lack of increase in MCV, fetalhemoglobin, or both, is not an indication to

discontinue therapy.

13. For the patient who has a clinical response, long-term hydroxyurea therapy is

indicated.

14. Hydroxyurea therapy should be continued during hospitalizations or illness. In case

Patient doesn’t respond to Hydoxurea crizanlizuamb may be given to older patients

16 and above. Crizanlizumab is only recommended to the patients who are

experiencing severe adverse events when on Hydoxurea theraphy. Or on patients on

maximum dose of Hydoxurea but are stillexperiencing more than three VOCs per

annum

15. New data suggests lower doses of hydroxyurea may also be beneficial. Titrate to

response.

References:

Page 227: Training Manual for Hemoglobinopathies and Hemophilia:

227

1. Alli NA, Patel M Recommendations for the management of sickle cell disease in

South Africa. S Afr Med J. 2014 Nov;104(11):743-51.

2. Barbara P. Yawn, George R. Buchanan Management of Sickle Cell Disease Summary

of the 2014 Evidence-Based Report by Expert Panel Members. JAMA September 10,

2014 Volume 312, Number 10

3. http://www.nhlbi.nih.gov/health-pro/guidelines/current/management-sickle-cell-

disease.htm (Accessed on September 30, 2014).

4. John J Strouse. Is low dose hydroxyurea the solution to the global sickle cell crisis?

Pediatric Blood cancer 2015 June:62(6)929-30

Exchange transfusion and simple transfusion in Sickle Cell Disease

All patients with Sickle cell disease may not have anemia.

Avoid blood transfusion unlessanemia is symptomatic.

Transfusion therapy not indicated for management of uncomplicated pain crisis.

Patient should receive an annual check- up at level 3 or 4 center with an expert

or at anytime if there are complications.

Blood transfusion is a supportive therapy in Sickle cell disease

Blood transfusion therapy in SCD can be used regularly (prophylactically) to prevent

or manage complications like stroke or only in the case of an acute life threatening

complications.

Effect of blood transfusion in SCD patients due to-

o Dilution of HbS red blood cells by normal containing cells from normal

donors.

Page 228: Training Manual for Hemoglobinopathies and Hemophilia:

228

o Decrease in percentage of HbS, due to the longer circulating lifespan of HbA /

normal red blood cells.

o Increase in Hb oxygen saturation levels by approximately 1 to 6 %, this results

in increased oxygen delivery to tissues.

o Suppression of erythropoietin.

SCD patients are more likely to have complications of blood transfusion like-

alloimmunization (antibody to donor minor blood group antigens), febrile non-

hemolytic transfusion reactions etc.

BEFORE starting regular transfusion support consider performing or sending patient

for extended blood grouping.

Alloimmunization to red cell antigens is a common occurrence in transfused patients

with SCD. Alloimmunization occurs rapidly after the initiation of transfusions

without extended matching.

Alloimmunization to minor RBC antigens in patients with SCD can be associated

with vaso-occlusive episodes and life-threatening hemolysis. Equally important,

alloimmunization can result in a greater risk of future transfusion reactions and

greater difficulty identifying compatible units of blood

Strong recommendation for those patients with clinically significant antibody

inthepast to get matching for minor RBC antigens (C, E, and Kell) to reduce the

risk of alloimmunization,

DNA typing may be done at higher centers. genotyping can be performed after

transfusions, if needed, to determine sources of incompatibility.

Other precautions of any transfusion in SCD patients-

Use of prestorage leukodepletion or white blood cell filter to decrease the rate of

febrile non-hemolytic transfusion reactions.

Individuals with SCD should not receive blood transfusion therapy from

individuals with sickle cell trait as part of regular transfusion therapy program.

No evidence of PRBC Storage time linkage with outcomes.

Irradiation not required unless planning for BMT.

Immunize against hepatitis B.

Monitor for iron overload and start chelation therapy appropriately.

Page 229: Training Manual for Hemoglobinopathies and Hemophilia:

229

Simple transfusion

Uses of simple blood transfusion — Simple blood transfusion involves transfusion of one or

more units of blood without removal of the patient's blood.

Table 24 Simple transfusion in sickle cell disease

Indications of simple transfusion Benefit of simple transfusion

1 Severe anemia (ie, Hb <5 g/dL), Increases oxygen carriage capacity, lowers

Sickle Hemoglobin (HbS) concentration,

without increasing blood viscosity

2 Preoperative transfusion to reduce

complications of surgery

Based on clinical trial data, lowers Sickle

Hemoglobin (HbS) concentration

3 Use of simple blood transfusion therapy

in individuals who have a Hb <10 g/dl

with symptoms of anemia

Increases oxygen carriage capacity, lowers

Sickle Hemoglobin (HbS)

When simple transfusion may not be useful.

Blood hyper viscosity: Blood viscosity is an interaction of several parameters-

Hemoglobinlevel,percentageof sickle hemoglobin (HbS) blood flow rate, and white

blood cell count.

High blood viscosity beyond normal threshold results in low oxygen delivery.

Hyperviscosity is a serious problem and though its symptoms are non-specific and related,

centralnervoushyperviscosity can cause acute neurologic injury, and can be associated with

central venous thrombosis or cerebral infarcts.

Contraindications of Simple

transfusion

Risks of simple transfusions

1. Patients with SCD Relative risk- may cause hyperviscosity syndrome in

The types of blood transfusion are- simple and exchange.

Care needed to avoid complications related to hyperviscosity or transfusion

which increasesthehemoglobin above 12g/dl.

Page 230: Training Manual for Hemoglobinopathies and Hemophilia:

230

complications whose

hemoglobinlevel is near

baseline (7 to 9 g/dL)

children or adults with SCD because it raises the

Hemoglobin and reduces percentage of HbS to a lower

level.

Viscosity is strongly affected by Hb S concentration.

Monitor AND recheck at Level 3, 4 Center

2 Avoid use of simple

blood transfusion

therapy in individuals

who have a Hb

>10 g/dL and a HbS

percentage >50 percent

of total Hb.

High Risk of hyper viscosity

Exchange blood transfusion

The other type of transfusion support is exchange transfusion. Exchange blood transfusion

involves removing some amount of the patient’s blood along with giving PRBC transfusion.

There are both automated apheresis and manual methods of performing this procedure.

Table 25: Comparison of Simple and Exchange transfusion in sickle cell disease

Comparison of simple versus exchange transfusion

Comparative reductioninSicklehemoglobin

in simple versus exchange transfusion

Baseline HbS levels in individuals with

SCD are at least 90 % of total

hemoglobin and after simple transfusion

this goes down to >75%

In an exchange transfusion the

percentage of sickle Hb can be reduced

to lower levels (<30 %)

Simple transfusion leads to iron overload more

rapidly, while with exchange transfusion there

isless need for iron chelation.

Less need for chelation with exchange

transfusion

Preoperative setting, randomized trials have found

simple transfusion to be equivalent to exchange

transfusion in preventing perioperative

Equal

Page 231: Training Manual for Hemoglobinopathies and Hemophilia:

231

complications

Exchange transfusion preferred over simple

transfusion-(evidence)

Acute stroke (Strong)

Severe acute chest syndrome (Strong)

Multi-organ failure (Strong)

Preoperative transfusion with baseline Hb > 10

g/dL (Moderate)

Transfusion of volume sensitive patients

(Moderate)

Refractory priapism (Uncertain)

Exchange transfusion better

Though no randomized clinical trials have been done to compare simple versus exchange

transfusion; clear clinical indications and benefits of exchange transfusion have been seen in

SCD with suspected stroke, respiratory failure, and multi-organ failure situations.

Table 26: indications of Exchange transfusion in sickle cell disease

Indications of Exchange transfusion Benefit

Multi-organ failure,

Suspected stroke,

Respiratory compromise,

Acute chest syndrome

Management of acute emergency.

Rapid and more reduction in HbS

concentration.

Only exchange transfusion can significantly

lower HbS levels to <30 %.Aim for 15-20%

HbS level to minimize sickling

complications.

Reduced blood viscosity. Improved blood

flow

Regularly scheduled exchange transfusions

used in the prevention of stroke, acute chest

syndrome, and recurrent painful episodes

Some patients with severe phenotype and

without anemia, may require this. Follow

advice of level 2 or 3 center.

Exchange transfusion therapy can be full blood volume exchange or partial volume exchange

and the method may be manual or automated apheresis. A full exchange transfusion rapidly

lowers the HbS level to 30 % or less and improves the hemoglobin level. In general,

Page 232: Training Manual for Hemoglobinopathies and Hemophilia:

232

automated apheresis is preferred over manual exchange because it can be done faster and

causes fewer volume shifts. However both are effective in reducing Hb S levels.

Table 27: Comparison of Partial and full volume exchange transfusion in sickle cell diseas

Partial exchange/ limited Full blood volume exchange transfusion

Partial exchange transfusion refers to a

limited exchange transfusion that is less

effective in lowering the HbS level but is

more easily performed. In order to lower the

HbS below 30%, repeat partial exchange

transfusions may be necessary.

A full exchange transfusion allows for

rapid lowering of the HbS level to 30 % or

less, and correction of anemia

Easier to perform Needs technical skill and experience to

manage fluid shifts

With acute organ deterioration, such as respiratory failure, stroke, or multi-organ failure,

we suggest lowering the HbS level into the range of 15 to 20 %, and raise total Hb to 10 to

12 g/dL.

In critically ill patients this will minimize sickling complications.

HbS level is unlikely to increase above 30 % within four weeks, so less need for repeat

exchange till that time.

Blood transfusion volume

For acute sickle cell complications, the dose of red cells should be calculated to result in

a Hb 10 g/dL or HCT 30%.The volume required for simple and exchange transfusions are

estimated by the patient weight and hematocrit; extra care needed for children. Avoid wasting

blood for transfusion, round up to closest unit for adults, discuss with blood bank for making

smaller bags

● Children : a transfusion of 10 mL/kg will increase the Hb 2.5 to 3.0 g/dL

Page 233: Training Manual for Hemoglobinopathies and Hemophilia:

233

● Adults : each unit of PRBC will increase the Hb concentration by approximately

1 g/dL

1. Formula used for estimation of simple transfusion volumes:

Packed RBC volume for

SIMPLE transfusion (mL) = ([dHCT - iHCT] x TBV) ÷ RpHCT

[dHCT is the desired hematocritiHCT is the initial hematocrit (both given as percent;eg-

40percent) ];

[TBV is the estimated total blood volume in mL (e.g., 60 mL/kg in adult women,

70 mL/kg in adult men, 80 mL/kg in children, 100 mL/kg in infants)]

[RpHCT is the hematocrit of the replacement packed RBC (55 to 60%)].

2. Formula used for estimation of EXCHANGE transfusion volumes

Manual partial

exchange volume (mL) = ([dHCT - iHCT] x TBV) ÷ (RpHCT - [(iHCT + dHCT) ÷ 2])

[dHCT is the desired hematocritiHCT is the initial hematocrit (both given as percent;eg- 40

percent)]

[TBV is the estimated total blood volume in mL (e.g., 60 mL/kg in adult women,

70 mL/kg in adult men, 80 mL/kg in children, 100 mL/kg in infants)].

[RpHCT is the hematocrit of the replacement packed RBC (55 to 60%)].

Table 28: over transfusion management in SCD

In case hemoglobin level increases over 10g/dl, in patients after transfusion

in symptomatic SCD patients, necessary action

Use phlebotomy for all patients with a post-

transfusion Hb ≥13.0 and HbS>50%.

To reduce hyper viscosity

If a patient'shemoglobin has been raised to

above 12 g/dL

with simple transfusions and the HbS levels

are >50 % of total hemoglobin,

use phlebotomy to decrease the hemoglobin

to 10 g/dL

To reduce hyper viscosity

In adults the maximum amount of blood phlebotomized should not exceed two units

Page 234: Training Manual for Hemoglobinopathies and Hemophilia:

234

At any point of time consultation with level 3 or level 4 center is indicated for patient safety

and best management.

References

1. Ted Wun, Kathryn Hassell. Best practices for transfusion for patients with sickle cell.

Disease. Hematology Reviews 2009; volume 1:e22

2. Koehl B, Sommet J, Holvoet L, et al. Comparison of automated erythrocytapheresis

versus manual exchange transfusion to treat cerebral macrovasculopathy in sickle cell

anemia. Transfusion 2016; 56:1121.

3. Savage WJ, Reddoch S, Wolfe J, Casella JF. Partial manual exchange reduces iron

accumulation during chronic red cell transfusions for sickle cell disease. J

PediatrHematol Oncol 2013; 35:434.

4. Charache S. Treatment of sickle cell anemia. Annu Rev Med 1981; 32:195.

5. Carrara P, Balocco M, Pinto V, et al. Manual erythroexchange for chronic transfusion

therapy in patients with sickle cell syndromes unresponsive to hydroxyurea: a long-

term follow-up. Am J Hematol 2010; 85:974.

6. Nieburg PI, Stockman JA. Rapid correction of anemia with partial exchange

transfusion. Am J Dis Child 1977; 131:60.

7. Casella JF, King AA, Barton B, et al. Design of the silent cerebral infarct transfusion

(SIT) trial. PediatrHematol Oncol 2010; 27:69.

Page 235: Training Manual for Hemoglobinopathies and Hemophilia:

235

Management of newborn and pregnancy

Newborn screening

Rationale for newborn screening

The greatest chance of dying in sickle cell disease is the first year of life. Several of these

causes of early death may be prevented or more effectively treated. Prophylaxis and

education may only be put in place if the underlying diagnosis is known.

Technique of newborn screening:

• HPLC followed by DNA based tests

• Another option is isoelectric focussing.

Management

• Confirm diagnosis on capillary sample at age 1 month; once confirmed,children

should be registered in a sickle cell clinic for regular follow-up, education and

counselingofthefamily.

• Offer screening tests for parents to allow counseling on future risks of affected

children.

Page 236: Training Manual for Hemoglobinopathies and Hemophilia:

236

• Ensure regular immunization schedules (sickle cell patients often miss immunizations

because of transient fevers, illness), including hemophillus influenza type B vaccine.

• Teach parents how to feel for the spleen which should be done daily more often if the

child is sick.

• Advice purchase of thermometer and teach its use.

• Penicillin prophylaxis.

• Explain about delayed physical and sexual development and slim build, advice plenty

of fresh fruit and vegetables and advice against expensive tonics to fatten the child

• Arrange regular review of child or immediately if sick.

• Stress importance of attendance when well in order to establish steady state blood

levels and clinical features.

• Provide walk in facilities or immediate contact opportunities if sick.

• Routine monitoring at all visits –The following are essential elements of all visits

totheSCD clinician:

• A complete blood count (CBC) and reticulocyte count.

• Height, weight and head circumference

• Vital signs including blood pressure

• Room air oxygen saturation

• Extended red blood cell antigen typing.

Table 29

Counseling required in care of infant/child with sickle cell disease

AGE TOPICS

1 month Confirmation of hemoglobinopathy diagnosis, introduction to

sickle disease, preliminary education regarding prophylactic

penicillin administration and fever management.

2-3 months Review of hemoglobinopathy genotype and family studies;

discussion of sickle cell pathophysiology; discussions

regarding immunizations, penicillin prophylaxis, and fever;

initial education regarding splenic sequestration.

5-6 months Review of adherence with penicillin prophylaxis, education

about spleen palpation, provision of information about therapy

options [hydroxyurea, chronic transfusion, bone marrow

transplant] and research.

Page 237: Training Manual for Hemoglobinopathies and Hemophilia:

237

8-9 months Review of adherence with penicillin prophylaxis, review of

spleen palpation, discussion about dactylitis and pain events

12 months Discussion about acute chest syndrome, discussion about

hydroxyurea [and potential initiation of treatment ]

Pregnancy

SCD causes polymerisation of the abnormal hemoglobin in low-oxygensituations. This

causes it to become rigid and take on the fragile sickle-shaped red cells. These cells

causevaso-occlusion in the small blood vessels, leading to acute painful crises. stroke,

pulmonary hypertension, renal dysfunction, retinal disease, leg ulcers, cholelithiasis and

avascular necrosis. These cells are also likely to breakdown and result in hemolysis and

anemia. Women with sickle cell disease have increased complications for themselves and

their babies.

What are the additional risks to the woman and the baby?

Baby: Increased incidence of perinatal mortality, fetal growth restriction.

Mother:Premature labour and acute painful crises during pregnancy.

Some studies have shown increase in spontaneous miscarriage, maternal mortality,

infection, thromboembolic events and antepartum haemorrhage. Also,there are some

reports of increased risk of pre-eclampsia and pregnancy-induced hypertension .

What should be checked for women planning pregnancy?

When planning for pregnancy in women with SCD ensure they have received and are

compliant with basic SCD care and medications, immunizations , prophylactic

penicillin, folic acid etc.

They should have their routine assessment for complications of sickle cell disease at

their level 2 center and be referred to level 3 or 4 center if there are complications of

disease.

Screening for pulmonary hypertension with echocardiography, this may be associated

with increased mortality.

Blood pressure and urinalysis should be performed to identify women with

hypertension and/or proteinuria. Renal and liver function tests should be performed

annually to identify sickle nephropathy and/or deranged hepatic function.

Page 238: Training Manual for Hemoglobinopathies and Hemophilia:

238

Screening for iron overload, by serum ferritin, in multi-transfused patients,T2*

cardiac magnetic resonance imaging may be helpful to assess iron overload in heart

and liver. If high iron overload is aggressive,iron chelation before conception is

advisable.

Screening for red cell antibodies. Red cell antibodies may indicate an increased risk of

haemolytic disease of the newborn.

What test is required for the father?

If the mother has sickle cell disease or is a carrier for any hemoglobinopathy/ variant

hemoglobin trait, then the father should also be tested. If he is normal then likelihood

of baby being affected withaserioushemoglobin disorder is less (though silent

mutations cannot be ruled out).

If father is also a carrier of a hemoglobinopathy /variant hemoglobin trait, the genetic

counseling required and prenatal testing option to be given for serious disorders.

What medicines should be stopped?

Preconceptually Hydroxyurea should be stopped at least 3 months before conception.

Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers should

be stopped before conception.

Antenatal care

All routine antenatal tests and care are required.

Regular antenatal checkups required

Blood pressure and urinalysis should be performed at each consultation and

midstream urine for culture performed monthly.

Additionally

o influenza vaccine should be recommended.

o Women with SCD should aim to avoid precipitating factors of sickle cell

crises such as exposure to extreme temperatures, dehydration and

Page 239: Training Manual for Hemoglobinopathies and Hemophilia:

239

overexertion. Persistent vomiting can lead to dehydration and crisis.If this

occurs, should receive iv fluids.

o Iron supplementation should only be given if there is laboratory evidence of

iron deficiency.

o Low-dose aspirin 75 mg once daily from 12 weeks of gestation to reduce the

risk of developing pre-eclampsia.

o Women with SCD may be advised to receive prophylactic low-molecular-

weight heparin during antenatal hospital admissions.

o Non-steroidal anti-inflammatory drugs (NSAIDs) should be prescribed only

between 12 and 28 weeks of gestation owing to concerns regarding adverse

effects on fetal development.

Ultra-sound examination

o Offer a viability scan at 7–9 weeks of gestation.

o Offer routine first-trimester scan (11–14 weeks of gestation) and a detailed

anomaly scan at 20 weeks of gestation.

o May require serial fetal biometry scans (growth scans) every 4 weeks from 24

weeks of gestation.

Blood transfusion during pregnancy?

Though routine prophylactic transfusion is not recommended, if a woman with SCD

develops complications then an exchange transfusion is required. Such patients may

even require repeat transfusions every 4 weeks during pregnancy.

Blood should be matched for an extended phenotype including full rhesus typing (C,

D and E) as well as Kell typing. During pregnancy ensure leukodepletion of all blood

products.

Table 30:Indications for transfusion of blood during pregnancy in sickle cell disease.

Women who are on a chronic transfusion

regimen before pregnancy , continue to

prevent disease complications

Continue same transfusion regimen

Acute chest syndrome Exchange transfusion

Twin pregnancy Simple/ Exchange transfusion

Anemia- simple transfusion Simple transfusion

Page 240: Training Manual for Hemoglobinopathies and Hemophilia:

240

Women with previous serious medical,

obstetric or fetal complications

Exchange transfusion

How to manage acute pain crisis during pregnancy?

This is the commonest cause of admission with 27% and 50% of women having a painful

crisis during pregnancy. Advice to avoid excess heat/ cold environment, exercise,

dehydration is Tablerequired. If it occurs then-

Admit to the hospital or give daycare management under supervision.

A multidisciplinary team should assess for diagnosis of sickle pain crisis or other

medical complications, acute chest syndrome (ACS), sepsis or dehydration.

Urgent fluids

If pain crisis give analgesia within 30 minutes of reaching the hospital - The World

Health Organization analgesic ladder should be used, starting with paracetamol for

mild pain, for severe pain may need opioid. s (avoid pethidine in SCD patients -risk

of seizures).

o NSAIDS - if 12–28 weeks of gestation.

o Weak opioids such as Tramadol/codeine can be used for moderate

pain, and stronger opiates such as morphine can be used for severe

pain.

o If requiring opioids –admit

o Morphine can be given by the oral, subcutaneous, intramuscular or intravenous route

depending on the woman’s preference and local expertise.

o Parenteral opiates can be given by intermittent bolus or patient-

controlled administration systems.

o With opiods-Prescribe laxatives, antipruritic and antiemetic

o Monitor pain, sedation, vital signs, respiratory rate and oxygen

saturation every 20–30 minutes until pain is controlled and signs are

stable , then every 2 hours. Thromboprophylaxis should be given to

women admitted to hospital with acute painful crisis.

Type of delivery and timing?

Pregnant women with SCD with normal growing fetus- vaginal delivery after 38+0

weeks of gestation.

Page 241: Training Manual for Hemoglobinopathies and Hemophilia:

241

SCD is not considered a contraindication to attempting vaginal delivery. Obstetric

advice for c-section needed.

Blood should be cross-matched for delivery. Good hydration maintained at all times.

Ideally delivery should take place in center familiar with sickle cell disease patients.

Monitor for SCD complications in post partum time as well. Low-molecular-weight

heparin should be administered while in hospital and 7 days post-discharge following

vaginal delivery or for a period of 6 weeks following caesarean section.

What anesthesia to be offered to women with SCD?

Should have pre-anesthesia evaluation in third trimester of pregnancy.

Avoid the use of pethidine, though other opiates are safe to use.

Recommended to use Regional analgesia for caesarean section.

References

1. Afolabi BB, Iwuala NC, Iwuala IC, Ogedengbe OK. Morbidity and mortality in sickle

cell pregnancies in Lago, Nigeria: a case control study. J

ObstetGynaecol2009;29:104–6.

2. Rajab KE, Issa AA, Mohammed AM, Ajami AA. Sickle cell disease and pregnancy in

Bahrain. Int J GynaecolObstet2006;93:171–5.

3. National Institute for Health and Clinical Excellence. Hypertension in pregnancy.

The management of hypertensive disorders during pregnancy. NICE clinical guideline

107. London: NICE; 2010.

4. Howard RJ, Tuck SM, Pearson TC. Pregnancy in sickle cell disease in the UK: results

of a multicentre survey of the effect of prophylactic blood transfusion on maternal and

fetal outcome. Br J ObstetGynaecol 1995;102;947–51

5. Management of Sickle Cell Disease in Pregnancy. Green–top Guideline No. 61 July

2011.https://www.rcog.org.uk/globalassets/documents/guidelines/gtg_61.pdf

(accessed 6 May 2019)

Page 242: Training Manual for Hemoglobinopathies and Hemophilia:

242

Alloimmunization

All Multi-transfused patients are at risk of RBC allo-immunization i.e. they develop

antibodiestocertainRBC antigens. Though higher in patients of sickle cell disease and non

transfusion dependent thalassemia (NTDT), but may occur in any patient. Allo-immunization

puts these transfusion recipients at risk of hemolytic transfusion reactions if the antibody is

not detected at the time of pre-transfusion testing. Allo-immunization also shrinks the

precious donor pool for the allo-immunized patient as he can receive blood only from donors

who are negative for the antigen against which the recipient has developed antibodies.

Patients who develop one antibody are usually responders and have a propensity to develop

more allo-antibodies, this further shrinks the eligible donor pool sometimes making

compatible blood unavailable when required.

Page 243: Training Manual for Hemoglobinopathies and Hemophilia:

243

Allo-immunization should be suspected when pre-transfusion testing shows incompatibility

or the expected rise in hemoglobin is not seen and other causes for it are ruled out. A rise in

hemoglobin may be seen in some recipients but it is followed by a rapid fall in the levels.

Factors Impacting Alloimmunization

Rates of alloimmunization vary according to demography and heterogeneity of the

population group. Several contributing factors for alloimmunization are:

1. Responder Status

Every individual has a unique genetic makeup which makes them vulnerable to

several serological problems after chronic transfusion therapy. This genetic difference

helps us divide individuals into three broad categories:

Non-Responders – These are the individuals who never develop any allo antibodies

despite repeated exposures.

Responders – These are the individuals who develop at least one alloantibody with

one or more exposures.

Hyper-Responders – These are the individuals who develop multiple allo antibodies

with one or more exposures.

2. Antigenic [RBC and human leukocyte antigen (HLA)] disparity between the

blood donor and the recipient which will increase further if they both belong to

different ethnic groups.

3. Genetic Factors: Although the patient could produce antibodies against both red cell

antigens and/or Human leucocyte antigens separately after exposure to foreign

antigens, yet according to few researchers, formation of antibodies against blood

group antigens, for example, Rh, Kell, Kidd, Duffy, Diego could be accentuated

because of certain HLA phenotype of the individual. Thus some HLA phenotypes

make individuals more prone to alloimmunization making them responders or hyper-

responders.

4. Presence of leucocytes in Donor PRBCs/Platelets: Donor leucocytes are mediators

of direct antigen presentation. Leucodeplteted blood components (BCs) cause less

alloimmunization as compared to non-leucodeplted BCs.

5. Antigenic Dose: The higher the antigen dose, the greater is the risk of allo-

immunization.

Page 244: Training Manual for Hemoglobinopathies and Hemophilia:

244

6. Underlying Disorder: Persons with sickle cell anemia/HbD are at a higher risk of

alloimmunization as compared to patient cohorts.

7. Number of transfusions received: As the total number of BCs transfused increases,

so does the risk of alloimmunization.

8. Age at first transfusion: Starting PRBC transfusions at an early age confers some

immunotolerance and reduces the overall risk of alloimmunization.

9. Immunogenicity of antigen: Rh system antigens – D, C, E, c, and e and Kell antigen

K are the most immunogenic and most alloimmunized patients develop antibodies to

these RBC antigens. Transfusing PRBCs which are matched for extended Rh and K

phenotype has been proposed as a mechanism to reduce alloimmunization.

Implications of Alloimmunization:

1. Increased risk of delayed hemolytic transfusion reaction (DHTR) or delayed

serological hemolytic transfusion reaction (DSTR).

Allo-antibodies against cellular antigens or blood group antigens might disappear

from detection over time. If antigen re-exposure during subsequent transfusions

occurs, it can induce an anamnestic antibody response resulting in delayed

hemolytic transfusion reactions which could become life-threatening.

2. Decreased chances of finding a compatible blood product for future use thus

increasing turn-around time of blood. If alloantibody is detectable during

compatibility testing procedure, then antigen negative blood component unit has

to be found and this becomes more difficult if more than one alloantibodies are

present. Sometimes the alloantibody is masked by the presence of auto-antibodies

thus difficult to detect.

3. It will decrease post-transfusion survival of RBCs in the recipient. As the antibody

present in the recipient will cause lysis and thus the destruction of transfused

donor RBCs.

4. It might accelerate iron loading of tissues and organs as a higher number of RBCs

need to be transfused. Chronic transfusion and underlying inflammatory state of

Page 245: Training Manual for Hemoglobinopathies and Hemophilia:

245

the recipient cause elevations of the pro‐inflammatory cytokines TNF‐α and IL‐2

thus further accentuating iron overloading in these patients.

Approaches to Mitigate Risk of Alloimmunization:

1. Donors of similar ethnic descent should be considered to decrease the chances of

genetic disparity and hence alloimmunization.

2. Use of fresh blood (within one week of the collection): Due to metabolic and

rheological alteration of RBCs older blood units are associated with post-

transfusion hemolysis, decreased survival and alloimmunization.

3. Preventive phenotyping of the patient: Before starting transfusion therapy of

patients with hemoglobinopathies, ABO forward and reverse grouping along with

Rh and Kell typing should be done. It would be helpful in identifying antibodies

in case alloimmunization occurs.

4. Antibody screening could be performed using a sensitive method as a part of

routine pre-transfusion testing.

5. Leukoreduction of the blood components: Pre-storage or bedside leukoreduction

approaches could be helpful in preventing alloimmunization.

Management of Alloimmunization:

1. Antibody Screening using a three cell panel of reagent red cells with known

extended phenotypes ( commercially available) each time a patient's sample is

received.

2. In case the patient is antibody screen positive, the patient's plasma/serum

should be tested against an 11 cell panel of reagent RBCs of known extended

phenotypes( commercially available) to identify the antibody specificity.

3. Once the antibody specificity has been identified, donor RBCs should be typed

for the antigen of interest and antigen negative units should be selected for the

patient.

4. The antigen negative units should be cross-matched with the

recipient'splasma/serum and only cross-match compatible antigen negative

units should be issued for transfusion.

5. Antigen negative units should be provided for previously documented

antibodies even if the antibodies are undetectable at the time of antibody

Page 246: Training Manual for Hemoglobinopathies and Hemophilia:

246

screening/identification and cross-match in order to prevent immune HTRs

due to an anamnestic response.

6. Extended Rh and Kell antigen matched PRBCs should be provided when

possible especially in patients who have developed one or more alloantibodies.

Problems encountered:

1. Majority of blood banks have limited donor pools i.e. there is a lack of sufficient

phenotypically matched donors to provide blood to allo-immunized patients.

2. As patients of hemoglobinopathies are candidates of long term blood transfusion

therapy they might receive blood from different hospitals during their lifetime.

Different hospitals have different serological testing approaches for patients for

example, not all the institutes are providing phenotypically matched blood to their

patients and also some institutes are doing extended phenotype matching for all

the patients with hemoglobinopathies while others are only using for the patients

who have developed alloantibodies.

3. Undue delays could be faced by the patients in case we are not able to find

phenotypically matched blood for the patient and/or compatibility testing is

complicated by the presence of autoantibody.

4. Providing extended antigen-matched blood unit even in alloantibody negative

individuals might cause logistics issue at some centers.

References

1. Kruatrachue M., Sirisinha S., Pacharee P., Chandarayingyong D., Wasi P. An

association between thalassemia and autoimmune haemolytic anemia (AIHA)

Scandinavian Journal of Haematology. 1980;25(3):259–263.

2. Singer ST, Wu V, Mignacca R, Kuypers FA, Morel P, Vichinsky EP.

Alloimmunization and erythrocyte autoimmunization in transfusion-dependent

thalassemia patients of predominantly Asian descent. Blood 2000; 96: 3369–3373.

3. Spanos T, Karageorga M, Ladis V, et al. Red cell alloantibodies in patients with

thalassemia. Vox Sang 1990;58:50-5.

4. Datta SS, Mukherjee S, Talukder B, Bhattacharya P, Mukherjee K. Frequency of red

cell alloimmunization and autoimmunization in thalassemia patients: a report from

eastern India. Adv Hematol 2015; 2015:610931

Page 247: Training Manual for Hemoglobinopathies and Hemophilia:

247

5. Gehrie EA, Tormey CA. The Influence of Clinical and Biological Factors on

Transfusion-Associated Non-ABO Antigen Alloimmunization: Responders, Hyper-

Responders, and Non-Responders. Transfus Med Hemother. 2014;41(6):420–429.

6. Yazdanbakhsh K, Ware RE, Noizat-Pirenne F. Red blood cell alloimmunization in

sickle cell disease: pathophysiology, risk factors, and transfusion management. Blood.

2012;120(3):528–537.

7. Gehrie EA, Ness PM, Bloch EM, Kacker S, Tobian AAR. Medical and economic

implications of strategies to prevent alloimmunization in sickle cell disease.

Transfusion. 2017;57(9):2267–2276. doi:10.1111/trf.14212

8. Bao W, Zhong H, Li X, et al. Immune regulation in chronically transfused allo-

antibody responder and nonresponder patients with sickle cell disease and β-

thalassemia major. Am J Hematol. 2011;86(12):1001–1006.

Hematopoietic Stem Cell Transplantation(HSCT) for Hemoglobinopathies

Hematopoietic Stem Cell Transplant for Thalassemia Major

Learning Objectives:

A. To discuss the need and anticipated outcomes of Hematopoietic Stem Cell Transplant

(HSCT) in Thalassemia major

B. Understand the risk categorization for patients undergoing HSCT and to recognize a

very high risk group.

C. Highlight the need of doing HSCT at a young age and preferably before the age of 7

years.

D. Address source of stem cells and alternative stem cell donor related issues in HSCT

for Thalassemia major

E. To understand the transplant procedure/protocol (that includes pre transplant work up,

preparative regimen and post-transplant follow up)

A]Need for HSCT in Thalassemia major:

Currently, an allogeneic hematopoietic stem cell transplant (HSCT) remains the only curative

option for patients with β thalassemia major. The use of HSCT is rapidly increasing in India

and other developing countries and is hence the most widely available and accessible curative

Page 248: Training Manual for Hemoglobinopathies and Hemophilia:

248

therapeutic strategy for this condition. The central concept revolves around the ability to

replace the hematopoietic stem cells (HSC) from a donor to a recipient resulting in a new

donor derived hematopoietic system in the recipient. Significant advances over the last two

decades have resulted in a steady improvement in clinical outcomes for patients with this

disorder undergoing such a procedure. Currently in patients with good risk features it is

reasonable to anticipate a greater than 90% chance of a successful transplant outcome. Even

among those with high risk features, success rates are approaching 80%. These improvements

have resulted from use of better risk stratified conditioning regimen and more effective

supportive care. With thalassemia being a significant public health problem in the country,

there is need for capacity building and increasing the centers capable of offering this therapy.

B] Pre-transplant risk assessment and stratification of patients with Thalassemia

major:

Risk stratification of patients with β thalassemia major undergoing a myelo-ablative

allogeneic (SCT) classifies them into three risk groups (Pesaro Class I, II and III) based on

liver size (>2cm), presence of liver fibrosis and inadequate iron chelation. Patients with none

of the above risk factors are classified as Class I, those with one or two of these risk factors

are Class II while those patients who have all three adverse risk factors are classified as Class

III (summarized in Figure 9). Patients in Class I and II our considered to be low risk and have

an excellent long term outcome following an allogeneic SCT. Class III patients on the other

hand are considered high risk and have inferior outcomes following a SCT. However, in a

population with poor medical treatment prior to SCT, the above risk stratification is limited

by its failure to recognize the significant heterogeneity among patients in Class III. The

Pesaro risk stratification does not recognize a very high risk subset of Class III in these

populations perhaps because such patients hardly exist in Western countries. With allo-SCT

being increasingly offered in many developing countries where this category of patients exist

in large numbers, recognizing this high risk group is extremely important to suitably modify

their HSCT protocols. Additionally, it must be stressed that HSCT is contraindicated in

patients who already have evidence of severe end organ damage from high iron overload

leading to organ dysfunction such as cardiac failure of cirrhosis, such patients even if

considered for HSCT should only be done in centers with substantial experience and

expertise with these procedures.

Figure 9: Risk factor and risk groups on pre-transplant evaluation of patients with

Thalassemia major undergoing an HSCT

Page 249: Training Manual for Hemoglobinopathies and Hemophilia:

249

C] Importance of doing HSCT at a young age and preferably before the age of 7 years:

Retrospective analysis of factors impacting clinical outcome on large data set from India

analyzed in 2007, helped identify age ≥7 years old and had a liver size ≥5 cm in a patient pre-

transplant to constitute what has been shown to be a very high risk subset of a conventional

Class III group (Class III high risk)[2]. The adverse impact of age (≥7 years) and liver size (>

2 cms) was further validated by an international collaborative analysis in 2010. The

significance of this differentiation has been emphasized by the fact that outcomes of HSCT

were clearly different in these two groups when treated with the same protocols. Class III and

more specifically Class III high risk subset have a high risk of graft rejection and regimen

related toxicity (RRT), especially sinusoidal obstruction syndrome (SOS) leading to multi-

organ failure and death. These complications are perhaps related to the high degree of allo-

immunization and iron over load related end organ damage in this cohort. When such a

population is transplanted even in a developed country with expertise in such transplants the

rejection rate is as high as 34%.

D] Source of stem cells and alternative stem cell donors for HSCT in Thalassemia

major:

Bone marrow (BM) has been the preferred choice of stem cells in non-malignant

hematological disorders to reduce the risk of GVHD, though the incidence of both acute and

chronic GVHD in this predominantly pediatric population is low. Peripheral blood stem cell

grafts (PBSC) when used have been reported to be associated with faster engraftment and

lower requirement of blood product support in the peri-transplant period and have also been

associated with a low incidence of graft rejection.

Page 250: Training Manual for Hemoglobinopathies and Hemophilia:

250

The best stem cell donor is a HLA identical sibling donor unfortunately, only about a third of

patients with thalassemia major have a HLA matched sibling donor which limits access to an

HSCT for these patients. There is a rare possibility of another related donor, such as a parent,

grandparent or cousin, being HLA identical though the prior probability of this is extremely

low.

Matched unrelated donors:Use of a matched unrelated donor (MUD) SCT has the potential

to overcome this. The initial results with this approach, prior to the advent of high resolution

HLA typing, were dismal with a 55% graft rejection. Since then, there has been significant

improvement in MUD SCT with high resolution molecular HLA typing becoming standard in

MUD selection. Outcome in malignant disorders with MUD is now comparable to that of

HLA matched sibling transplants. More recent data suggests that OS of 79% can be achieved

with this approach with thalassemia free survival of 66% and a 25% chance of treatment

related mortality [16]. This study also suggested that an extended haplotype match was

associated with a superior outcome. The overall data would suggest that MUD SCT should

only be considered in centers that have reasonable experience with this approach and is best

limited to low risk patients at present. High resolution HLA typing with a full (10 of 10)

match is the preferred donor and in addition they should ideally not have HLA-DP1

mismatches in the direction favoring graft rejection.

Cord blood transplants: Cord blood transplants over the last decade have been the most

rapidly growing source of stem cells for allogeneic SCT. However, for patients with

thalassemia major there is limited data. Using cord blood stem cells for allogeneic SCT in

thalassemia major must be considered under two categories, which are distinctly different in

terms of clinical outcome:

1. Related cord blood transplant: when a HLA matched or partly mismatched sibling is

the source of stem cells

2. Unrelated cord blood transplant: when an unrelated cord blood product is procured

from a cord blood bank as part of donor search for an allogeneic SCT. There could be

varying degrees of HLA mismatch.

Using related cord blood the Eurocord Transplant Group reported a 2 year probability of

thalassemia free survival of 79% in 33 patients with thalassemia.

Unrelated cord blood transplant genuinely increases the potential pool of donors for patients

with thalassemia major. Unfortunately the published data is limited. In a recent review over 6

Page 251: Training Manual for Hemoglobinopathies and Hemophilia:

251

studies a total of 19 patients were reported. After combining data from three different

registries, Ruggeri et al reported in 2011, that the cumulative graft failure rate was an

unacceptable 52%. At this time point one cannot recommend an unrelated cord blood

transplant outside the setting of a clinical trial.

Haplo-identical stem cell transplants:There is a lot of interest in haplo-identical stem cell

transplants in the world over the last few years. Novel conditioning and GVHD prophylaxis

regimens have resulted in dramatic improvements in clinical outcome even without T cell

depletion of the graft. There is however very limited data in thalassemia major. In one small

series (n=22) using T cell depletion grafts the graft rejection rate was 27% and the TFS about

67%. More recently the use of grafts with depletion of CD3αβ T cells looks promising with a

few successful reports. In a disease where several management options exist and newer ones

are on the horizon, whether a treatment option that gives less than ~80% thalassemia free

survival can or should be recommended needs further discussion. Haplo-identical SCT should

therefore, at this point of time, be done preferably in the setting of a clinical trial and only in

tertiary care centers.

E] Transplant procedure:

Prior to initiating the procedure a detailed pre-transplant evaluation of patient and donor is

done to confirm diagnosis and HLA match status and also to exclude any condition that is a

contraindication to proceeding with a transplant, in both the recipient and the donor. The

conventional conditioning regimen that is used is a combination of busulfan and

cyclophosphamide. More recently a number of novel conditioning regimens have been

introduced and popular alternative is a treosulfan based conditioning regimen that replaces

busulfan and is better tolerated with less adverse events. The first report on the use of

treosulfan being used as part of the conditioning regimen for thalassemia, in a small series of

20 patients of which 45% were Class III and 18 were matched unrelated stem cell transplants,

was by Bernado et al. Treosulfan based conditioning regimen is ideally suited for patients

with thalassemia major including very high risk patients. The low hepatic and other toxicity

profile over a wide range of AUCs obviates the need for drug dose monitoring and makes this

an attractive agent to use in the conditioning regimen.

Post-transplant patients need to be monitored carefully for engraftment. Patients continue to

remain under high risk for infections for the first year. An additional complication that can

Page 252: Training Manual for Hemoglobinopathies and Hemophilia:

252

occur in a proportion of cases and can occasionally be fatal includes graft rejection and graft

versus host disease, neither of which one can accurately predict.

The iron overload state continues to require attention post transplantation. Post a successful

transplant provided the patients is stable and his Hb is above 10mg/dl, then the preferred

method of iron removal is phlebotomy. It can be repeated once in 14 days and a volume of

6ml/kg can be removed in one sitting. If the hemoglobin level is not adequate or if

phlebotomy is not possible then the patient should be started on chelation therapy. It should

be continued (maybe required for years) till the ferritin level is <100ng/ml. The optimal

pharmacological agent(s) and chelation regimen post-transplant remains to be defined. In

addition to chelation these patients need close attention to immunization, endocrine and organ

dysfunction secondary to iron overload.

References:

1. Mathews, V., A. Srivastava, and M. Chandy, Allogeneic stem cell transplantation for

thalassemia major. Hematol Oncol Clin North Am, 2014. 28(6): p. 1187-200.

2. Mathews, V., et al., A new stratification strategy that identifies a subset of class III

patients with an adverse prognosis among children with beta thalassemia major

undergoing a matched related allogeneic stem cell transplantation. Biol Blood

Marrow Transplant, 2007. 13(8): p. 889-94.

3. Lucarelli, G., et al., Bone marrow transplantation in patients with thalassemia. N

Engl J Med, 1990. 322(7): p. 417-21.

4. Lucarelli, G., et al., Bone marrow transplantation in adult thalassemia. Blood, 1992.

80(6): p. 1603-7.

5. Hongeng, S., et al., Outcomes of transplantation with related- and unrelated-donor

stem cells in children with severe thalassemia. Biol Blood Marrow Transplant, 2006.

12(6): p. 683-7.

6. Fang, J.P. and L.H. Xu, Hematopoietic stem cell transplantation for children with

thalassemia major in China. Pediatr Blood Cancer, 2010. 55(6): p. 1062-5.

Page 253: Training Manual for Hemoglobinopathies and Hemophilia:

253

7. Sabloff, M., et al., HLA-matched sibling bone marrow transplantation for beta-

thalassemia major. Blood, 2010. 117(5): p. 1745-50.

8. Phrommintikul, A., et al., Splenectomy: a strong risk factor for pulmonary

hypertension in patients with thalassaemia. Heart, 2006. 92(10): p. 1467-72.

9. Chiesa, R., et al., Unpredictability of intravenous busulfan pharmacokinetics in

children undergoing hematopoietic stem cell transplantation for advanced beta

thalassemia: limited toxicity with a dose-adjustment policy. Biol Blood Marrow

Transplant, 2010. 16(5): p. 622-8.

10. Chandy, M., et al., Randomized trial of two different conditioning regimens for bone

marrow transplantation in thalassemia--the role of busulfan pharmacokinetics in

determining outcome. Bone Marrow Transplant, 2005. 36(10): p. 839-45.

11. Rajasekar, R., et al., Cellular immune reconstitution and its impact on clinical

outcome in children with beta thalassemia major undergoing a matched related

myeloablative allogeneic bone marrow transplant. Biol Blood Marrow Transplant,

2009. 15(5): p. 597-609.

12. Mathews, V., et al., Improved clinical outcomes of high risk beta thalassemia major

patients undergoing a HLA matched related allogeneic stem cell transplant with a

treosulfan based conditioning regimen and peripheral blood stem cell grafts. PLoS

One, 2013. 8(4): p. e61637.

13. Iravani, M., et al., Comparison of peripheral blood stem cell transplant with bone

marrow transplant in class 3 thalassemic patients. Exp Clin Transplant, 2010. 8(1): p.

66-73.

14. Ghavamzadeh, A., et al., Peripheral blood versus bone marrow as a source of

hematopoietic stem cells for allogeneic transplantation in children with class I and II

beta thalassemia major. Biol Blood Marrow Transplant, 2008. 14(3): p. 301-8.

15. Gaziev, D., et al., Bone marrow transplantation from alternative donors for

thalassemia: HLA-phenotypically identical relative and HLA-nonidentical sibling or

parent transplants. Bone Marrow Transplant, 2000. 25(8): p. 815-21.

16. La Nasa, G., et al., Unrelated donor bone marrow transplantation for thalassemia:

the effect of extended haplotypes. Blood, 2002. 99(12): p. 4350-6.

17. Fleischhauer, K., et al., Graft rejection after unrelated donor hematopoietic stem cell

transplantation for thalassemia is associated with nonpermissive HLA-DPB1

disparity in host-versus-graft direction. Blood, 2006. 107(7): p. 2984-92.

Page 254: Training Manual for Hemoglobinopathies and Hemophilia:

254

18. Pinto, F.O. and I. Roberts, Cord blood stem cell transplantation for

haemoglobinopathies. Br J Haematol, 2008. 141(3): p. 309-24.

19. Ruggeri, A., et al., Umbilical cord blood transplantation for children with

thalassemia and sickle cell disease. Biol Blood Marrow Transplant, 2011. 17(9): p.

1375-82.

20. Bertaina, A., et al., HLA-haploidentical stem cell transplantation after removal of

alphabeta+ T and B-cells in children with non-malignant disorders. Blood, 2014.

21. Bernardo, M.E., et al., Treosulfan-based conditioning regimen for allogeneic

haematopoietic stem cell transplantation in patients with thalassaemia major. Br J

Haematol, 2008. 143(4): p. 548-51.

Hematopoietic Stem Cell Transplant for Sickle Cell Disease

A.1.0 Need for HSCT in Sickle Cell Disease (SCD):

Sickle cell disease is the most common inherited hemoglobinopathy worldwide, and is a

devastating, life threatening disease with limited therapeutic options to reduce disease

Learning Objectives :

A. To discuss the need and outcome of Hematopoietic Stem Cell Transplant(HSCT) in sickle

cell disease (SCD)

B. To identify the candidates suitablefor transplant in SCD

C. To understand the transplant procedure/protocol (that includes pre transplant work up,

preparative regimen and post-transplant follow up)

D. To identify the warning signs in post-transplant patients.

E. To discuss special issues in HSCT for SCD

Page 255: Training Manual for Hemoglobinopathies and Hemophilia:

255

severity.Although more than 94% of children with SCD in well-resourced countries now

survive until the age of 18 years due to routine newborn screening, penicillin prophylaxis,

primary stroke prevention, and hydroxyurea (HU) therapy, chronic complications (that

include stroke, sickle lung disease, renal failure, RBC allo-immunization, etc.) severely

impact the quality of life (QoL), and mortality is still significant once patients reach

adulthood. Majority of patients develop end organ damage due to repeated sickling crisis

episodes, which ultimately severely cripple their young adult life. In India, the scenario is

even more dismal. Despite the fact that the sickle phenotype prevalent in our country is of

milder form as compared tothe African counterpart due to high HbF levels, it is estimated that

about 20 per cent of children with sickle disease died by the age of two as reported in one

ICMR survey, and 30 per cent children with SCD among the tribal community die before

they reach adulthood.1 Additionally as SCD is particularly prevalent in scheduled populations

in our country, which comprise the most socioeconomically disadvantaged communities with

lack of medical facilities to care for their disease, the estimates for chronic complications for

SCD are expected to be very high.2

Thus it is estimated that unlike inthewest, majority of

patients of SCD who reach adulthood in our country develop chronic complications, thereby

significantly compromising their QoL as well as adding liability to the already burdened

health care system of our country. In light of this, allogenic stem cell transplant is the only

treatment strategy that can actually cure this disease. All other treatment options (like HU

therapy, regular transfusion therapy) for the disease only ameliorate the disease severity

rather than actually curing it.

A.1.1 Transplant Outcome in Sickle Cell disease:

HSCT should be considered standard of care when a patient has an indication and an HLA-

identical sibling donor. The first successful cure of SCD after HSCT was reported in

a single pediatric patient in 1984 who had SCD and coexisting acute myeloid leukemia.3

Since then, over 1,000 patients have received an HLA-identical sibling HSCT worldwide

with a 5-year Event Free Survival (EFS) and Overall Survival (OS) of 91.4% and 92.9%,

respectively.4 EFS is lower with increasing age at transplantation and higher for

transplantations performed after 2006 given improvements in preparative regimens,

supportive care, and management of complications. The data on transplant outcome in SCD

from India is sparse, the reasons for which are numerous. Firstly, majority of patients

requiring Allo-SCT for their disease are either not referred to higher centre in a timely

Page 256: Training Manual for Hemoglobinopathies and Hemophilia:

256

manner or they do not have a matched sibling allogenic donor (found in only 10% of patients)

to undergo transplant. Additionally as previously mentioned, that the hot spot areas for the

severe variety of the disease requiring transplant is predominantly located in the tribal areas

of Central India and Orissa where the patients are very poor to afford this treatment. However

with the Government’s renewed interest in the prevention & treatment of this disease at the

national level, the financial roadblock for the transplant may be overcome in the near future.

B.1.0 Indications for Stem cell transplant in Sickle cell disease:

• Stroke or central nervous system event lasting longer than 24 h

• Impaired neuropsychological function with abnormal cerebral MRI scan

• 3 or more episodes per year of acute chest syndrome leading to recurrent

hospitalizations in patients on Hydroxyurea (HU) therapy for at least 9 months

• More than 3 episodes per year of vaso-occlusive crisis requiring hospitalizations in

patients on HU therapy for at least 9 months

• Stage I or II sickle lung disease (patients with pulmonary hypertension but

without/minimal limitation of physical activity)

• Sickle nephropathy (moderate or severe proteinuria defined as urinary protein to

creatinine ratio of >50 mg/mmol (442 mg/g) or a glomerular filtration rate 30 to 50%

of the predicted normal value)

• Bilateral proliferative retinopathy with major visual impairment in at least one eye

• Osteonecrosis of multiple joints

• Red-cell alloimmunization during long-term transfusion therapy

• TricuspidRegurgitantJetvelocity≥2.7 m/son2D-Echo

• RegularRBCtransfusiontherapy(≥8 transfusionsperyear. for ≥ 1 y)toprevent. vaso-

occlusivecomplications.

B.1.1 Contraindications for Stem cell transplant in Sickle cell disease:

• Karnofsky or Lansky functional performance score <50-70

• Acute hepatitis with evidence of intrahepatic cholestasis or cirrhosis on biopsy

• Severe renal impairment (GFR<30ml/min/1.73m2)

• Severe cardiac disease

Page 257: Training Manual for Hemoglobinopathies and Hemophilia:

257

• Stage III or IV sickle lung disease (Patients with pulmonary hypertension resulting in

marked limitation of physical activity).

• Demonstrated lack of compliance with medical care

B.1.2 Timing of Allogeneic SCT in Sickle cell Disease:

Young patients’preferably preschool age (2-5 yrs)with symptomatic SCD who have an HLA-

matched sibling donor should be transplanted as early as possible. This strategy is often

beneficial as it prevents permanent end organ damage, thereby also improving the transplant

outcome and quality of life.

B.1.3 Source of stem cell :

Unmanipulated Bone Marrow/Peripheral blood stem cell (PBSC)

C.1.0 Pre transplant Evaluation :

• History & Physical Examination

• Any pre-exisiting Co-morbidities

Routine :

• Complete Blood Count (CBC), Kidney Function Test (KFT), Liver Function

Test (LFT) , Urine Routine Examination

• Coagulation Screen (PT/APTT/Fibrinogen)

• Viral Serology: Hepatitis B, C ,HIV, CMV, EBV

• S. Ferritin (Quantitative)

• Surveillance Cultures (Blood, Urine, Stool. Throat swab)

• Chest X Ray

• HLA typing (High Resolution HLA-A,B,DQ, DR)

To Assess End Organ Damage:

• CNS: Cerebral magnetic resonance imaging (MRI) and magnetic resonance

angiography

• Pulmonary :Chest X Ray; PFT ((total lung capacity, forced vital capacity,

residual volume, and the ratio of forced expiratory volume to forced vital

Page 258: Training Manual for Hemoglobinopathies and Hemophilia:

258

capacity), HRCTindicated only in case of strong clinical suspicion of Sickle

lung disease

• Hepatic: Liver biopsy/R2 MRI if S.Ferritin>2500ng/ml to assess hepatic iron

overload

• Opthalmic : Fundus Examination (to r/o retinopathy)

• Renal: Urine protein/creatnine ratio, 24 hr. urine protein (to r/o proteinuria)

• 2-D Echo: to r/o pulmonary Hypertension

C.1.1. Preparative/Conditioning Regimen:

• Myeloablative. conditioning regimen comprising of myeloablative doses of Busulfan

(14-16 mg/kg in divided doses administered over 4 days); Cyclophosphamide (200

mg/kg in divided dosesadministered over 4 days) along with immunosuppressive

doses of Rabbit ATG (10-15mg/kg given over 3 days) with BM/PBSC as the HSC

source has resulted in excellent OS (91-100%) & EFS (82-100%) in several studies.5

The addition of ATG decreases the riskof graft rejection from 22.6% to 3% and

should be considered as standard of care in HSCT myeloablative preparative

regimens.6 Methotrexate (on day +1.+3,+6,+11) along with cyclosporine (maintaining

trough levels 200-300 ng/ml) is recommended as standard Graft versus Host Disease

(GVHD) prophylaxis. The overall rates of acute and chronic GVHD utilizing a

cyclosporine based immunosuppressive regimen range between 10-22%,though

GVHD was a main cause of treatment related mortality (TRM) in several

studies.7Prior HU therapy and young age at transplant are the strongest predictors of

successful treatment outcome.

C.1.2 Post Transplant Evaluation:

• Chimerism Studies – Quantitative restriction-fragment–length polymorphisms (RFLP)

or tandem repeats in DNA on Day 30, 60, 90 & 365.

• Cerebral MRI with angiography: at 180 and 365 dayspost transplantation.

• Hypertension monitoring at each OPD visit

• Iron Overload (S. Ferritin)

• Osteoporosis/ Avascular Necrosis

Page 259: Training Manual for Hemoglobinopathies and Hemophilia:

259

• Pulmonary function test (Annually)

• Renal: Urine Protein/ Creatinine Ratio, 24 Hr Urine Protein (Annually)

• Ophthalmic Examination (Annually)

To be monitored annually

• 2nd Malignancy

• Hypogonadism

• Dyslipidemia

• Thyroid function

Vaccinations : Inactivated or killed vaccines in all eligible patients between 6 and 12

months after transplantation

D.1.0 Warning Signs in Post transplant patients:

The presence of any of the following symptoms/signs in a post-transplant patient may suggest

GVHD/any serious infection and warrant urgent admission or referral to higher centre.

• Fever > 100.5 F lasting for >24 hrs.

• Loose motions

• Redness/rash over the skin

• Persistent vomiting

• Dryness of mouth/eyes

• Ulcerations in the mouth/ difficulty in swallowing

• Significant weight loss

• Recurrent fall in haemoglobin level

E.1.0 Special issues during HSCT in SCD:

• Seizures:It is one of the most dreaded complications during/after stem cell transplant

particularly in SCD patients. The etiology may be multifactorial that include

intracerebral haemorrhage, vasculopathy, hypoxic injury due to sickling, uncontrolled

hypertension or hypomagnesemia/electrolyte disturbances. Anticonvulsant

prophylaxis with phenytoin/levetiracitam should be routinely prescribed to all the

patients and continued for six months post transplantation. In addition other

supportive measures like intensive monitoring of blood pressure, magnesium levels

(maintained between 1.8-2.2 meq/dl), platelet count( maintained>20,000/cumm) and

haemoglobin levels (between 9-11gm/dl) should be done.

Page 260: Training Manual for Hemoglobinopathies and Hemophilia:

260

• Stable Mixed Chimerism (MC):Stable mixed chimerism with a reduction rather than

an elimination of hemoglobin S is sufficient to reverse the SCD phenotype as

erythropoiesis by a minority of engrafted donor cells can lead to a majority of

circulating normal erythrocytes with a survival advantage over short-lived sickle

RBCs. In these MAC regimens, donor chimerism values as low as 10% to 20% were

sufficient to improve Hb levels, HbS % and SCD-related complications.8

Younger age

(<10 years old), the use of ATG , were associated with more SCD patients achieving

mixed chimerism after HCT. Addition of ATG to the combination of busulfan and

cyclophosphamide increased the percentage of children with MC (50–95% of donor

cells) from 5% to 35% in children undergoing myeloablativeallo-HSCT, while

reducing the rejection rate.9

References

1. Serjeant GR, Ghosh K, Patel J. Sickle cell disease in India: A perspective. Indian J

Med Res. 2016;143:21-4.

2. Hockham C, Bhatt S, Colah R, Mukherjee MB, Penman BS, Gupta S, Piel FB.

Thespatial epidemiology of sickle-cell anaemia in India. Sci Rep. 2018; 8:17685.

3. Bernaudin F, Souillet G, Vannier JP, et al. Bone marrow transplantation (BMT) in 14

children with severesickle cell disease (SCD): the French experience. GEGMO.Bone

Marrow Transplant. 1993;12Suppl1:118-21.

4. Leonard A, Tisdale JF. Stem cell transplantation in sickle cell disease:therapeutic

potential and challenges faced. Expert Rev Hematol. 2018;11:547-565.

5. Glukman E, CapelliB, Bernaudin F, et al. Sickle cell disease: an international survey

of results of HLAidenticalsibling hematopoietic stem cell transplantation. Blood

2017:129:1548-56.

6. Walters MC, Patience M, Leisenring W, et al. Bone Marrow Transplantation for

Sickle Cell Disease. NEngl J Med. 1996 ;335:369–76.

7. Angelucci E, Matthes-Martin S, Baronciani D, et al. Hematopoietic stem cell

transplantation inthalassemia major and sickle cell disease: indications and

Page 261: Training Manual for Hemoglobinopathies and Hemophilia:

261

management recommendations from aninternational expert panel. Haematologica.

2014;99:811-20.

8. Walters MC, Patience M, Leisenring W, et al. Stable mixed hematopoietic chimerism

after bone marrowtransplantation for sickle cell anemia. Biol Blood Marrow

Transplant. 2001;7:665–73.

9. Hsieh MM, Kang EM, Fitzhugh CD, et al. Allogeneic hematopoietic stem-cell

transplantation for sicklecell disease. N Engl J Med. 2009 ;361:2309-17

Page 262: Training Manual for Hemoglobinopathies and Hemophilia:

262

Alpha Thalassemia: Pathophysiology and Diagnosis

Background:

Adult humans have the adult Hb (HbA) as their predominant hemoglobin (Hb), constituting

~97% of total Hb.The remainder is comprised of minor components of HbA2 (2-3.5%) and

fetalHb (HbF, <1%). The Hb molecule contains 4 polypeptide globin chains associated with 4

heme groups. Synthesis of the globin chains, defects in which forms the basis of the

thalassemias, requires two globin genes - α and β. The α-globin genes are duplicated and

located on the telomeric region of the short arm of chromosome 16 (16p13.3). These

duplicated α genes are present in a cluster which is shown in Figure 10 and contains an

embryonic α-like gene zeta (ζ), 3 pseudogenes (ψζ1, ψα1, ψα2), a theta (θ) gene of unknown

function along with two adult α-genes (α2 and α1). Expression of these α-globin genes is

regulated by sequences located in and around the structural genes and by aregion named HS-

40 that is located 40 kb upstream from the α-cluster. This region, contains an erythroid-

specific DNAase I hypersensitive site, multiple, small, highly-conserved binding sites for

trans-acting factors (NF-E2, GATA-1), several CACCC motifs and binding sites for YY1

transcription factors.

Figure 10: Spatial organisation of the human β- and α-globin gene clusters.

Disorders of globin chains can be classified into those associated with a quantitative

reduction in the polypeptide chain production, i.e. the thalassemias and those associated with

production of a varianthemoglobin like HbS, HbE etc., i.e. the hemoglobinopathies. A few

Page 263: Training Manual for Hemoglobinopathies and Hemophilia:

263

disorders show overlapping features and are designated thalassemichemoglobinopathies, for

e.g, HbLepore, HbE and Hb Constant Spring.

Thalassemias are further sub-classified based on the precise globin chain that is

underproduced, and thus include the α, β, δβ, δ, γ, γδβ, εγδβ and other rarer subtypes. This

manuscript deals exclusively with the α-thalassemias.

Nomenclature of α-Thalassemias

Alpha thalassemias can be α0 or α

+, deletional (-α) and non-deletional (α

T). Deletion or

inactivation of both the α-globin genes on one chromosome causes α0

thalassaemia. In the

heterozygous state, its genotype is represented as --/αα (heterozygous α0

thalassaemia) where

there is no output of α-globin from the affected chromosome. When only one of the two

linked α-genes (i.e. on the same chromosome 16) is inactivated, the condition is called α+

thalassaemia, and the genotype is written as -α/αα in cases in which one of the α-globin genes

is deleted, or αTα/αα if one of them is inactivated by a mutation.

Classification and Clinical Syndromes:Four phenotypic types of α-thalassemia can

occur, with loss of function of one to all 4 α-genes:

1) Single α-gene deletions, synonyms: silent carriers/ mild α-thalassemia/ heterozygous

α+ thalassemia (α-/αα): Individuals with a single defective α-globin gene, for e.g. the 4.2 kb

deletion (-α4.2

), are asymptomatic and can only be identified on DNA analysis. Neonates

show a mild increase (1-2%) in Hb Bart’s (γ4 tetramers), a feature that can be exploited for

screening.

2) Two α-gene deletions, i.e. either homozygous α+ thalassemia (α-/α-), or heterozygous

α0 thalassemia (--/αα): Here, the quantitative reduction of α-protein is greater. Individuals

with this condition have red cell microcytosis, hypochromia and erythrocytosis. Most remain

asymptomatic and are detected during family screening. There can however be mild anemia

with levels of Hb Bart’s in the range of 5-6%.

3) Hemoglobin H Disease (α-/--): In HbH diseasepatients, only one out of four α-globin

genes is functional. The magnitude of reduction of α-protein is however, highly

heterogeneous but may lead to moderate to severe anemia (TI phenotype), sometimes with

splenomegaly, bone deformities and chronic fatigue. HbH is formed due to excess β chains

(β4 tetramers).

Page 264: Training Manual for Hemoglobinopathies and Hemophilia:

264

4) Hb Bart’s HydropsFetalis or α-thalassemia major (--/--): Complete absence of α-globin

genes leads to a marked relative excess of γ-globin in the fetus. The abnormal Hb molecule

(γ4 tetramers) is called HbBart's. Patients with HbH disease die in utero or shortly after birth.

Both HbH and Bart’s are non-functional Hbs with very high oxygen affinity. Women

carrying a fetus with Hb Bart’s hydropsfetalis have an increased rate of pregnancy-related

hypertension and hydramnios and an increased rate of peripartum haemorrhage and retained

products of conception.

Interaction of the α-globin genotype with β-thalassemia: Several ethnic groups with a high

prevalence of β-thalassemia also have a high frequency of deletional α-thalassemia; hence

patients who have co-inherited both of these conditions are not uncommon. Homozygotes or

compound heterozygotes for β-thalassemia who co-inherit α-thalassemia have lesser excess

of free α-globin chains and may have less severe symptoms. While co-inheritance of a single

α-gene deletion does not have a remarkable effect on the β-thalassemia phenotype,

individuals with two α-gene deletions coinherited with homozygous β0/β+ thalassemia often

exhibit a mild form of β- thalassemia intermedia (TI). Patients whoco-inherit HbH disease

(i.e. have only one functioning α-gene) along with homozygous β-thalassemia also have a TI

phenotype.

Conversely, increased α-globin product in β-thalassemia trait (βTT) further exacerbates the

globin chain imbalance, converting a clinically asymptomatic βTT to βTI with worsening of

clinical severity. The effect of increased α-globin chain production by two extra α-globin

genes in homozygous triplicated α-globin genes (ααα/ααα) or heterozygous quadruplicated α-

globin genes (αααα/αα) individuals is clearly demonstrated in β-thalassemia heterozygotes,

by resulting in TI phenotype.

Genetic Mechanisms and Pathophysiology:

Deletional α-thalassemia: Molecular pathology of the common alpha globin gene deletions

is depicted in Figure 11. The α2 and α1 globin genes are embedded within two 4 kb

homologous units which have homologous sub-segments X, Y, Z. The Z segments are 3.7 kb

away and reciprocal recombination results in only one α-gene (rightward deletion -α3.7

) and

the other allele has three α-genes (ααα3.7

). Recombination between the X boxes which are 4.2

kb apart results in leftward deletion -α4.2

and an ααα4.2

allele.

Page 265: Training Manual for Hemoglobinopathies and Hemophilia:

265

Several variable length deletions completely abolish both α-genes leading to α0 alleles.

Deletions of upstream regulatory elements which leave the alpha globin genes intact can also

cause α0-thalassemia. This phenomenon in patients has shown that HS-40 (MCS-R2) is

essential as a regulatory element possibly due to recombinations between the rich Alu repeats

and subtelomeric rearrangements.

Figure 11: Duplicated X, Y, Z box arrangement containing the alpha genes and

crossovers leading to deletions and triplications.

Non-deletional types of α-thalassemia: Unlike β-thalassemias, point mutations are

uncommon in α-globin genes. The first non-deletional form was described in 1977 and since

then >70 mutations have been described. The overwhelming majority affect the α2 gene and a

few are described in cis forms with deletions (-αT). These mutations act at various levels of

gene regulation and expression and include RNA splicing variants, frameshifts, nonsense

mutations, poly(A) signal variations and chain-termination mutations which result in

extension of alpha chains by 31 amino acids. Many of the variants lead to variably unstable

Hbs. Hb Constant Spring (αCS, α

142Gln) is an extended variant which is found in 4% of Thais.

Page 266: Training Manual for Hemoglobinopathies and Hemophilia:

266

Epidemiology of Deletional and Non-Deletionalα-Thalassemia:

High frequencies of α+deletional forms are found in the tropical belt, especially amongst

endogamous communities. α0deletions are most common in south-east Asia and the Eastern

Mediterranean islands but are only sporadically seen in the Indian subcontinent and the

middle-East. Common α0

deletions are --MED

in Mediterranean region and --SEA

in south-east

Asia. In north Indians, α+ thalassemia was first documented in patients of βTI by Garewal et

al. in 1994. Their study showed that 12-13% of the population had α+ thalassemia with

triplicated α-genes in 3%. The vast majorities (98%) are -α3.7

and only 2% are -α4.2

allele.

A study on 17 HbH patients from India showed that seven cases had the genotype --SA

/-

α3.7

while three had --SA

/--α4.2

and --SA

was found to be not uncommon in Indians. Non-

deletional α-thalassemias are common in south-east Asia and India. The predominant group

are the termination codon mutations which results in incorporation of 31 codons leading to an

elongated unstable α-chain. HbConstant Spring α142, Term→Gln (TAA>CAA in α2) is found

in frequencies of 5-8% in South East Asia. HbKoya Dora α142, Term→Ser (TAA>TCA in

α2) has been described.

Laboratory Diagnosis of α-deletions, point mutations and triplications

Symptomatic α-thalassemia should be considered after excluding common conditions such as

β-thalassemia syndromes. Iron deficiency anemia should be excluded, since it is commonly

encountered in most populations where both β- and α-thalassemia are common. Automated

blood counts (Hb and red cell indices) and peripheral smear examination reveal hypochromic

microcytosis and red cell anisocytosis (Figure 12). Reticulocyte counts and screening for

HbH inclusions using supravital staining should be done for suspected HbH disease (Figure

13). Hb HPLC is very useful to identify cases of HbH disease since twin pre-integration

peaks (Figure 14) are present. In addition, HbA2 levels are lower than normal (values <2%).

Hemoglobin electrophoresis at alkaline pH shows a fast-moving band after the adult Hb.

Tests for unstable Hbs (isopropanol and heat instability tests) are positive with HbH disease.

Red cell indices in alpha-thalassaemia: It is difficult to classify any particular individual

based only on red cell indices. Mean values of MCV and MCH are significantly lower in both

α+-thalassemia heterozygotes and homogygotes when compared to normal genotype. MCV

and MCH values are however not reliable to be used as good screen for α thalassemia in

isolation. Individuals with microcytic hypochromic anaemia and red cell indices suggestive

Page 267: Training Manual for Hemoglobinopathies and Hemophilia:

267

of β- thalassaemia traits or iron deficiency anemia should be investigated for α- thalassemia

after excluding β- thalassemia trait and iron deficiency by special tests.

Screening tests:

Demonstration of HbH Inclusion Bodies

Reagent: One % Azure Blue or New methylene blue. New batches of stain must be tested

with a known positive control because the redox action of the dyes may vary from batch to

batch.

Method:

1. Mix equal volume of fresh blood (within 24 hours of collection) with staining

solution.

2. Incubate at 37°C for 2 hours or at room temperature for 4 hours.

3. Resuspend the cells and spread a thin blood film after 30 min, 60 min, 90 min and 120

min. If inclusions are not seen at 2 hours, incubate overnight and re-examine before

labelling the test negative

4. Examine the film as for a reticulocyte count. The inclusion bodies appear as multiple

greenish-blue dots, like the pitted pattern on a golf ball (Figure 13).

They can be readily distinguished from reticulocytes, which exhibit uneven reticular

material or infrequent fine dots.

Interpretation & Comments

o In α+ thalassemia trait, only a very occasional HbH body (1:1000 to 1:10,000) is usually

seen; they are more numerous in α0 thalassemia,

o The number of cells developing inclusions is not reliable in differentiating the various

gene deletion patterns seen in α thalassemia

o Absence of demonstrable inclusions does not preclude a diagnosis of α thalassemia trait.

o This test is most useful in Hb H disease, where inclusions are usually found in more than

30% of red cells.

Page 268: Training Manual for Hemoglobinopathies and Hemophilia:

268

Figure 12: Peripheral blood findings in a patient with homozygous HbSallanches

[alpha2 104(G11) Cys>Tyr HBA2:c.314G>A)] showing marked hypochromic

microcytosis and anisopoikilocytosis.

Figure 13: “Golf ball” type HbH inclusions in red cells stained supra-vitally with

brilliant cresyl blue.

Page 269: Training Manual for Hemoglobinopathies and Hemophilia:

269

Figure 14: HPLC in a patient with HbH showing twin pre-integration peaks and

reduced HbA2 (BioRad Variant II™ Turbo Hemoglobin Testing System).

CE-HPLC Method: The Variant II™ Turbo HPLC instrument (Bio-Rad Laboratories,

Hercules, USA) applying the Beta-Thal Short™ Programmeis for quantitation of various

hemoglobin subtypes including adult hemoglobin (HbA), HbA2, fetalhemoglobin (HbF) and

any other abnormal hemoglobins. Five μl of whole blood required for this test undergoes

hemolysis at 12◦C and is sequentially injected into the analytical cartridge stream at 6.5-

minute intervals. Changes in absorbance are monitored in the eluate and displayed as a

chromatogram of absorbance versus retention time.

Data from the detector is processed by the built-in integrator software. Windows representing

characteristic ranges of retention time are used for result interpretation of the most

frequently-occurring hemoglobins. Retention time is defined as the time elapsed from sample

injection to appearance of the apex of the eluted peak. For hemoglobin fractions (i.e.HbA,

HbF and HbA2), the retention times, their proportions of the total hemoglobin (%) and the

peak characteristics are analyzed.

Page 270: Training Manual for Hemoglobinopathies and Hemophilia:

270

Cellulose Acetate Electrophoresis at Alkaline pH (pH 8.6):At alkaline pH,hemoglobin is

negatively charged. In an electric field it therefore migrates towards the anode (+). Structural

variants with surface charge difference will separate from HbA, however those without a

change in a charge will not. Cellulose acetate membranes are commercially available. They

contain about 80% air space in the form of pockets within the interlocking cellulose acetate

fibres. They come as dry, opaque, brittle films which crack easily if not handled properly.

When the film is placed in the buffer, the air spaces fill with liquid and then it can be easily

handled. These membranes can be made transparent for densitometry measurement of bands

using methanol and acetic acid mixture. The advantages of this technique are the speed of

separation (20-40 min) and the ability to store transparent membranes for longtime. Fully

automated instruments are now available for acid as well as alkaline pH electrophoresis

(Figure 15).

Figure 15. Automated Hb electrophoresis at alkaline pH (done on Genio S system,

Interlab, Rome) shows a fast moving relatively faint band of HbH in lane 2.

References:

1. Globin Gene Server: Laboratories of Computer Science, Engineering, Biochemistry

and Molecular Biology; Pennsylvania State University http://globin.cse.psu.edu

[accessed July 7, 2018]

2. Higgs DR, Weatherall DJ (2007) The alpha thalassaemias Cellular and Molecular Life

Sciences 1-9 DOI 101007/s00018-008-8529-9

3. Arnold SC, Quah TC, Low PS, Chong SS. A rapid and reliable 7-deletion multiplex

polymerase chain reaction assay for α-thalassemia Blood 2001; 98,250-251.

4. Garewal G, Fearon CW, Warren TC, et al. The molecular basis of beta thalassaemia

in Punjabi and Maharashtrian Indians includes a multilocus aetiology involving

triplicated alpha-globin loci Br J Haematol1994;86:372-6.

Page 271: Training Manual for Hemoglobinopathies and Hemophilia:

271

5. Nadkarni A, Phanasgaonkar S, Colah R, Mohanty D, Ghosh K. Prevalence and

molecular characterization of alpha-thalassemia syndromes among Indians Genet Test

2008;12(2):177-80.

6. Nadkarni AH, Gorakshakar AC, Sawant PM, et al. The phenotypic and molecular

diversity of hemoglobinopathies in India: A review of 15 years at a referral centerInt J

Lab Hematol 2019;41(2):218-226.

7. Sen R, Chakrabarti S, Sengupta B, De M, Haldar A, Poddar S, et al. Alpha-

thalassemia among tribal populations of Eastern India Hemoglobin 2005;29(4):277-

80.

8. Shaji RV, Eunice SE, Baidya S, et al. Determination of the breakpoint and molecular

diagnosis of a common alpha-thalassaemia-1 deletion in the Indian population Br J

Haematol2003;123:942-7.

9. Sharma P, Das R, Trehan A, et al. Impact of iron deficiency on hemoglobin A2% in

obligate β-thalassemia heterozygotes Int J Lab Hematol2015;37(1),105-11.

10. Trehan U, Garewal G, Kaul D, Das R. Alpha thalassemia and alpha gene triplications

in Punjabis, with and without beta thalassemia trait. Haematology 2000; 6,153-160

11. Purohit P, Dehury S, Patel S, Patel DK. Prevalence of deletional alpha thalassemia

and sickle gene in a tribal dominated malarial endemic area of eastern India ISRN

Hematol 2014;745254

Page 272: Training Manual for Hemoglobinopathies and Hemophilia:

272

Role of Genetic Modifiers

Genetic modifiers can impact the phenotypic severity of -thalassemia at the primary level by

affecting the degree of globin chain imbalance and at the secondary level by modulating

complications of the disease. Hence, a major determinant at the primary level is the deficit in

β-globin production(milder or severe β-globin gene mutation), and this relates to the nature

and number of copies of the -thalassemia alleles. This is then followed by the 2 major

secondary modifiers which are coinheritance of α-thalassemia and the innate ability to

produce fetalhemoglobin (HbF, α2γ2), both of which directly reduce the globin chain

imbalance.

Primary Modifier of Hemoglobinopathies:

The pathophysiology of β-thalassemia involves imbalance between α and β globin chains of

hemoglobin. (Figure 16) Thus the reduced amount or absence of beta globin chains results in

an excess of unbound alpha globin chains which precipitate in erythroid precursors in the

bone marrow and interfere with the maturation of the red blood cells leading to their

destruction in the bone marrow and subsequent anemia. Thus the primary modifier of the

disease severity is the type of β-globin gene mutation (point changes to small deletions) that

the patient inherits, which further determines the extent of down regulation of the β-globin

gene.Accordingly the β-globin gene mutations are classified as: βo

or null mutations which

lead to complete absence of the β-globin on the affected allele, β+

mutations which cause

residual production of beta globin chains and β ++

(mildor silent mutations) which lead to very

mild reduction in beta globin production. Depending on the type of the β-globin gene

mutations inherited, the patients present variable clinical phenotypes.Similarly in other β-

thalassemia syndromes: HbS-β thalassemia and HbE-β thalassemia, the severity depends on

the type of the β –thalassemia mutation inherited along with the variant hemoglobin.The

milder β –thalassemia mutations leading to amelioration of the disease severity in the Indian

population are -88 ( C→T), Cap site +1 (A→C) and Poly A (T→C). Figure 16 shows the

basic pathophysiology of thalassemia and the role of secondary modifiers.

Page 273: Training Manual for Hemoglobinopathies and Hemophilia:

273

Figure 16: Basic pathophysiology of β-thalassemia. B: Correction of globin chain

imbalance in presence of secondary modifiers of hemoglobinopathies

Secondary Modifiers of Hemoglobinopathies:

The search for secondary modifiers of hemoglobinopathies was accelerated by the

observation that though all sickle cell anemia patients have the same mutation in the β-globin

gene, the patients display a varying degree of clinical severity. Similarly in β-thalassemics, it

is often observed that patients with the same β-globin gene mutations have diverse clinical

phenotypes. This led to the identification of potential secondary modifiers of

hemoglobinopathies, which are as follows:

a. Co-inheritance of α-thalassemia:

In many populations where -thalassaemia is prevalent, -thalassaemia also occurs at a high

frequency and hence it is not uncommon to co-inherit both conditions. Homozygotes or

compound heterozygotes for -thalassaemia who co-inherit -thalassaemia will have a lesser

imbalance of these globin chains and tend to have a less severe condition. The degree of

amelioration depends on the severity of the β-thalassaemia alleles and the number of

functional α-globin genes.

Alpha-thalassemia is caused most frequently by deletions involving one or both alpha

globin genes. The most common deletions remove a single alpha globin gene, resulting in

the mild alpha+-thalassemia phenotype. However the co-inheritance of α-thalassemia in

hemoglobinopathy patients is often found to be associated with a milder disease phenotype.

In β-thalassemia syndromes, associated α-thalassemia minimizes the excess precipitation of

the α-globin chains and reduces the globin chain imbalance. Similarly in sickle cell anemia,

the co-existing α-thalassemia reduces intracellular sickle hemoglobin concentration, thereby

reducing HbS polymerization, sickling and thus decreasing the hemolysis.

Page 274: Training Manual for Hemoglobinopathies and Hemophilia:

274

In -thalassemia heterozygotes, increased -globin production tilts the globin chain

imbalance, converting a typically clinically asymptomatic state to that of thalassemia

intermedia. This is related to the co-inheritance of triplicated () or quadruplicated -

globin genes () instead of the number of globin genes (seen in normal

individuals.

Apart from the number of α-globin genes and an inherent capacity for producing -globin,

the proteolytic capacity of the erythroid precursors in catabolising the excess chainshas

often been suggested, but this effect has been difficult to define. Alpha hemoglobin

stabilising protein(AHSP), a molecular chaperone of α-globin has been suggested as another

genetic modifier but so far studies have been inconclusive.

b. Genetic modifiers of fetalhemoglobin:

All -thalassemias, heterozygous or homozygous conditions, have a variable increase in HbF,

which relates to the expanded erythroid mass and selective survival of the red blood cell

precursors that contain HbF.

The clinical pathogenicity of hemoglobinopathies develops during the first year of life when

the -globin genes are gradually silenced with the activation of δ and β-globin genes. Thus

continued -globin gene expression could be considered as a new therapeutic strategy to treat

hemoglobinopathies. Since an elevated fetalhemoglobin (HbF) level ameliorates the clinical

severity of the disease, identification of the potential genetic markers that raise the HbF levels

is very important.Asin β-thalassemics, the main cause of the disease is altered globin chain

ratio, the continued -globin gene expression leads to the production of more globin chains

which combine with the excess unbound α-globin chains, thus further reducing the globin

chain imbalance. Similarly in sickle cell anemia patients, raised HbF (α22) levels reduces

intracellular HbS polymerization. Both HbF and its mixed hybrid tetramer (α β γ) cannot

enter the deoxy sickle hemoglobin polymer phase, thus circumventing the cellular damage

evoked by HbS polymers. Recent studies have shown that in humans, the HbF level is

regulated by 4 major quantitative trait loci (QTLs): HBB locus region (11p15, mainly the -

globin promoter region), BCL11A (2p16), HBS1L-MYB intergenic region (6q23), KLF1

(19p13) which brings about 20-50 % variance in HbF levels.

Page 275: Training Manual for Hemoglobinopathies and Hemophilia:

275

HBB loci[-globin promoter]:

The elevated HbF levels in an adult are generally due to large HBB cluster deletions

which lead to Hereditary Persistence of FetalHemoglobin (HPFH ) , δβ thalassemia or due to

single point mutations in the -globin gene promoter. The probable mechanism of non-

deletional forms of HPFH is increased competition for the Locus Control Region (LCR) to

the mutant -globin gene promoter consequently increasing the -globin gene transcription,

with respect to the β-globin gene. Alternatively the variations in the promoter region, ablate

the binding of repressors or increase the attachment of enhancers, thus affecting the gene

expression. The most extensively studied and the first QTL for HbFvariation is the sequence

variant (C→T) at -158 position in the G-globin gene promoter that alters the Xmn I

restriction enzyme recognition site, (HBG2 c.-211 C→T). This variation is reported to

influence the G-globin gene expression under stress erythropoietic conditions. The T allele

of the XmnI polymorphism is found to be linked to raised HbF levels and is frequently

observed in thalassemia intermedia patients. Similarly, SCA patients with the Senegal or

Arab-Indian βS

haplotype which is linked to the T allele of the XmnIpolymorphism,also show

increased HbF levels as compared to patients with the Bantu haplotype (with thewild type C

allele) which showed lowest HbF levels and a more severe clinical course, thus suggesting

the role of the mutant allele in HbF induction. Other variations in the -globin promoter

region, like -117 (G>A), -175 (T>C), -198 (T>C) and -202 (C>G) also generate HPFH

condition, as they are located in functionally active sites of the -promoter region.

B-cell CLL / lymphoma 11 A [BCL 11 A]:

B-cell CLL/ lymphoma11A, a zinc fingertranscription protein encoded by theBCL11A gene

located in chromosome 2p16, plays a crucial role in postnatal development and normal

lymphopoiesis. Recent studies have shown that BCL11A is a direct repressor of γ-globin

gene expression in adult erythroid progenitors.BCL11A along with other hematopoietic

transcription factors (GATA1, SOX6, ZFPM1/FOG) and thechromatin remodelling complex

plays a critical role in repressing -globin gene expression. Genome wide association studies

(GWAS)have identified BCL11A genetic polymorphisms located in intron 2 to be associated

with higher fetalhemoglobin levels in hemoglobinopathy patients. It has been observed that

the raised HbF-associated region in intron 2 of BCL11A is a distinct regulatory sequence for

binding of RNA polymerase II and GATA-1 transcription factor both of which regulate gene

expression. The intron 2 region of BCL11A also shows Histone 3 acetylation thus suggesting

Page 276: Training Manual for Hemoglobinopathies and Hemophilia:

276

the role in gene expression. Thus, several polymorphisms in this regioneg the C allele of

rs11886868 (C>T), T allele of rs1427407 (G>T), variant allele A of rs4671393 (A>G ) are

associated with increased HbF levels in different population groups. These results indicate

the importance of this region in elevating the HbF levels.

Tertiary Modifiers: Complications of –thalassemia

These modifiers do not affect globin imbalance directly but might moderate complications of

the disease in many different ways. They include genetic variants which affect bilirubin

metabolism, iron metabolism, bone disease and cardiac complications. For example, jaundice

and a propensity to cholelithiasis is a common complication of -thalassemia and is attributed

to the rapid turnover of the red blood cells. Increased bilirubin levels and predisposition to

gallstones in β-thalassemia is associated with a polymorphic variant in the UGT1A1

promoter. Individuals who are homozygous for 7 [TA] repeats, also referred to as Gilbert

syndrome, have higher levels of bilirubin and increased predisposition to gallstones, an

observation that has been validated at all levels of β-thalassaemia. Several genes involved in

iron homeostasis have now been characterised, including those encoding hemochromatosis

(HFE), transferrin receptor 2 (TFR2), ferroportin (SLC4OA1), hepcidin (HAMP) and

hemojuvelin (HFE2). The H63D variant, a common polymorphism in HFE, appears to have a

modulating effect on iron absorption. β-thalassemia carriers who are homozygous for the

HFE H63D polymorphism, have higher serum ferritin levels than carriers without the

polymorphism. Figure 17 shows the diagrammatic representation of genetic modifiers of

hemoglobinopathies.

Thus, improved understanding of the influence of modifier genes involved in modulating the

complex pathophysiology of hemoglobinopathies may allow prediction of the disease

phenotype and promises to improve patient management and treatment.

Page 277: Training Manual for Hemoglobinopathies and Hemophilia:

277

Figure 17: Schematic representation ofgenetic modifiers of hemoglobinopathies

References:

1. Bank A. Regulation of human foetal haemoglobin: new players, new

complexities. Blood. 2006;107:435-43.

2. Rund D, Fucharoen S. Genetic modifiers in hemoglobinopathies. CurrMol

Med.2008;8:600-8.

3. Thein S. Genetic basis and genetic modifiers of β-Thalassemia and sickle cell disease.

Gene and cell therapies for beta-globinopathies. New York, NY: Springer New York;

2017:27-57.

4. Uda M, Galanello R, Sanna S, Lettre G, Sankaran V, Chen W, et al. Genome-wide

association study shows BCL11A associated with persistent foetal haemoglobin and

amelioration of the phenotype of β-thalassaemia. ProcNatlAcad Sci USA.

2008;105:1620-25.

Page 278: Training Manual for Hemoglobinopathies and Hemophilia:

278

Newer Modalities of Management for Hemoglobinopathies

Symptomatic hemoglobinopathies present either as thalassemia syndromes where

unbalanced alpha and beta chain synthesis leads to ineffective erythopoiesis as a major

cause of anemiaand iron overload. Other clinical presentations in different

hemoglobinopathies are due to specific structural hemoglobin defects leading either to

(a)Sickling disorders (b) Anemia (c) Methaemoglobinaemia(d) Polycythemia (e)

Intravascular hemolysis, etc.

Majority of symptomatic hemoglobinopathies are autosomal recessive in nature hence

need to be inherited from both parents either as a homozygous or double heterozygous

state. Cases of dominantinheritanceandclinical presentation due to unstable hemoglobins

or rare subsets of dominant thalassemias due to unstable globin chains are known.

Certain hemoglobinopathies like HbS-Oman produce symptomatic sickling state even in

hetereozygous state.

Hence modern management of hemoglobinopathies can be described as :

a) Prevention of thebirth of babies with symptomatic hemoglobinopathies by premarital,

prenatal genetic counseling and intervention

b) Transfusion practice to treat different hemoglobinopathies and advances made in this

area

c) Management of iron overload arising out of chronic transfusion and increased iron

absorption.

d) Management of infectious complications of chronic transfusion.

e) Prevention and management of chronic organ damage and management of sickle cell

disease.

f) Management of other clinical presentations of symptomatic hemoglobinopathies

g) Recent advances in management of Sickle cell anaemia and thalassemia

a) Prevention of new births of babies withserious symptomatic

hemoglobinopathies

Page 279: Training Manual for Hemoglobinopathies and Hemophilia:

279

For the last 50 years premarital/ marital genetic counselling forpreventing marriage/

pregnancy between two heterozygote individualsled to significant reduction in the birth of

thalassemic children from Cyprus, Sicily and other Mediterranean Countries .

Development of various molecular techniques toidentify foetuses with severe

hemoglobinopathies in heterozygous couples at risk by antenatal diagnosis has reduced

thebirth of new patients with symptomatic hemoglobinopathies. Such facilities are

available in 5-6 government centres in India and many private laboratories, corporate

hospitals in this country. Antenatal diagnosis should be associated with pre and post

diagnosis genetic counselling so that the couple can decide about the fate of the affected

pregnancy.

Other alternatives of adoption, artificial insemination by donor sperm and in some

advanced centres detection of thepaternal mutation in the foetus using non invasive

diagnostic procedures should be discussed.Advances in assisted reproductive technology

may allow preconceptional genetic diagnosis (PGD) and implantation of non affected

zygotes leading to anormal pregnancywhich is the most advanced form of preventive

management and today it may be available in a handful of private medical centres in this

country.

b. Advances in Transfusion Practice to manage symptomatic hemoglobinopathies

including thalassemia syndromes

Red cell transfusion forms one of the major foundations to manage severe anemia of

thalassemia and other hemoglobinopathies. This mode of management is also used to

manage various complications of commonly prevalent sickle cell syndromes.

Red cell transfusion can be given in several ways. However red cell transfusion is a

double edged sword and when chronically used it could be associated with several serious

complications like

i) Alloimmunisations against multiple red cell antigens & HLA antigens

ii) Certain serious transfusion reactions

iii) Transfusion transmitted viral and bacterial infection (HIV, Hep B, Hep C)

Campylobacter infection, Yersiniosis to name a few. (this increases many fold in

splenctomised cases)

iv) Transfusion associated iron overload and its consequences.

Page 280: Training Manual for Hemoglobinopathies and Hemophilia:

280

All the above mentioned complications could be minimized by optimizing and proper

schedulingof red cell transfusions using safe blood under good medical supervision.

Advances in this area includes

i) Using packed red cells for transfusion rather than whole blood( 4 mlof packed

redcells/kgincreases post transfusion hemoglobin by 1 g/dL) and maintaining pre

transfusion haemoglobin level at around 9 g/dLand frequency of transfusion of

once in 2-4 weeks for transfusion dependent thalassaemia major. Sickle cell

disease cases can tolerate lower haemoglobin levels better and keep it around 7-9

g/dL.

ii) Use of leucocyte removing filter (Filtration during blood donation is better than

filtration during bed side transfusion- both are available now). Leucodepleted red

cells produce less frequent alloimmunisation and substantially reduce some of the

transfusion reactions.

iii) The patient’s red cells should also be phenotyped (genotyped) for extended blood

group antigen at least ABO, Rh CcDEe, Kell, Duffy, Kidd & MNSs needs to be

typed. Effort should be made at least to match for ABO, RhDCcEe and Kell

blood group antigen. If that is not possible (It is not possible in most centres in

India now) then patients extended phenotype may help in gettingthe right kind of

red cells for transfusion when the patient develop alloantibodies.

iv) Strict records should be kept on amount of packed red cells required per year to

maintainthe pre-transfusion hemoglobin at 9 g/dl (normally in multitransfused

thalassemia patients this should not exceed 200ml/kg/year). If the requirement

increasesto>300ml/kg/yr then the patient should be assessed for hypersplenism

and alloantibody formation.

v) Microbiologically safest blood should be used for these patients i.e the red cells

should be obtained from regular non remunerated voluntary blood donors who

have no risk factor for viral infection during donation. The blood also should be

tested using both ELISA (4th

generation) or chemiluminescence assay along with

Nucleic Acid Amplification test (NAT test) for the three common transfusion

viral infections (HIV 1+2, Hep B & Hep C infection). The patient should be

regularly tested for these viral infections and appropriate management measures

should be taken if found positive.

Page 281: Training Manual for Hemoglobinopathies and Hemophilia:

281

vi) For certain complications of hemoglobinopathies like sickling crises, sickle cell

associated with pregnancy, sickle cell patients with stroke or pre –operative

preparation for such patients, partial red cell exchange transfusion may be

prescribed to bring down circulating Hb S levels to below 35-40% (As is found in

asymptomatic Hb S heterozygote patients). Similar technique may be adopted in

patients presenting with severe methaemoglobinemia due to haemoglobinopathies.

vii) Management of iron overload for chronic transfusion dependent thalassemia and

other hemoglobinopathies

Iron Overload is an important complication of chronic RBC transfusion therapy.

This is universally seen in patients with transfusion dependent thalassemia

patients. It is also seen in non transfusion dependent thalassemia intermedia

patients where gastrointestinal iron absorption is the major cause of iron overload.

Traditionally this complication needed continuous (10-14 hours/day)

subcutaneous infusion of desferiroxamine (40 mg-50 mg/kg/day) usingan infusion

pump. This modality of management was inconvenient and extremely costly.

Introduction of oral iron chelators like L1 (Deferiprone)( 70 mg/kg) and

Deferasirox-(FCT), Deferosirox (AsunraR, Desorox

R, Defriset

R). They are now

universally used orally at a dose of 30-40 mg/kg/day. The iron level is controlled

by regular (3 monthly) measurement of the dose of iron chelators to maintain a

serum ferritin between 500-1000g/L , Liver function renal function , serum

ferritin levels and complete blood counts should be regularly monitored. Target

seum ferritin levels should be around 1000 g/L.

d. Management of Infectious Complications of hemoglobinopathies

Chronic transfusion therapy increases the risk of HIV 1+2, Hep B and Hepatitis C infection

and use ofsafe blood and immunization against Hepatitis B before beginning transfusion

therapy is desirable. However once these infections are detected in the patient during regular

follow up, the treatment of such patients should be by experts in this field as newer

antiretroviral drugs as combination therapy are regularly evolving so also the therapy of

established chronic hepatitis B and C infection. If the patient is splenectomised, the chances

of infection due to pneumococcus, and hemophilus infection increase tremendously. Hence

Pneumococcal and Hemophilus vaccination must precede splenectomy and thepatient should

be on regular penicillin prophylaxis.Anyfever should be aggressively investigated and

treated.

Page 282: Training Manual for Hemoglobinopathies and Hemophilia:

282

e. Prevention and management of organ damage

Proper iron chelation may prevent damage to liver, heart and various endocrine organs. Iron

overload of the liver can synergistically work to aggravate liver damage. Regular check up

and replacement therapy in chronically transfused thalassemia patients with the hormones

from failing endocrine glands (Pituitary ,Thyroid, Pancreas, testis, ovary, supraenal,) may

allow proper growth and development in these patients.

Amerorrhoea, failure of ovulation and testicular failure are not uncommon so also pancreatic

diabetics and growth failure in transfusion dependent thalassemia as well as iron overloaded

non transfusion dependent thalassemia intermedia patients.

f. Management of other clinical presentation of symptomatic hemoglobinopathies

(i) All patients in this group presenting with anemia should receive adequate vitamins and

minerals including regular folic acid replacement.

(ii) Patients presenting with methaemoglobinaemia should receive adequate doses of

ascorbic acid. In emergency situation with this complication exchange transfusion and

intravenous methylene blue is indicated.

(iii) Patients presenting with polycythemia may require regular venesection to maintain PCV

around 45% for preventing consequences of hyperviscosity

(iv) Many patients with hemolyticanemia and ineffective erythropoiesis as a part of

symptomatic hemoglobinopathies may also have hyperuricaemia and need to be treated with

Allopurinol or Febustat.

(v) Patients with sickle cell syndromes i.e sickle cell disease, sickle thalassemia, sickle

hemoglobin – D disease may present with diverse complications due to sickling process and

may need specialist treatment. However the general rules to treat sickle cell syndromesare as

follows:

Adequate hydration

Pain relief

Folic acid 5 mg/daily

Complete all immunizations including Pneumococcal pneumonia, hemophilus

influenza and hepatitis B vaccine.

Substantial advances in sickle cell disease therapy have taken place now

Page 283: Training Manual for Hemoglobinopathies and Hemophilia:

283

a. Hydroxyurea between 10-25 mg/kg/ day was found to improve hemoglobin, reduces

number of painful and other crises.

b. Crizanlizumab (SEG 101) an anti P-Selectin humanized antibody significantly

prevents painful crisis in sickle cell disease patients at a dose of 5mg/kg given

intravenously once every 3-4 weeks.

Indications and Usage :

Crizanlizumab is indicated to reduce the frequency of vaso occlusive crises (VOCs) in

adult and pediatric patients aged 16 years and older with sickle cell disease

Recommended Dosage

Administer Crizanlizumab 5 mg/kg by intravenous infusion over a period of 30 minutes at

Week 0, Week 2, and every 4 weeks thereafter.

Administer Crizanlizumab diluted solution by intravenous infusion over a period of 30

minutes through an intravenous line, which must contain a sterile, nonpyrogenic 0.2 micron

inline filter.

vi. For various thalassemia syndromes

a. Hydroxyurea has been assessed as therapy to reduce number of blood transfusions and

improve hemoglobin levels.

70% of thalassemia intermedia patients responds to hydroxyurea therapy and can

come out of intermittent red cell transfusion therapy. However the results are not so

satisfactory with thalassemia major or symptomatic Hb-E thalassemia patients. Our

study has shown 30% of such patients on hydroxyurea significantly reduce their

transfusion requirements. Molecular studies at two centres showed homozygosity of

the Xmn1 polymorphism(+/+) and co-inheritance of alpha thalassemia improves the

chances of positive results with hydroxyurea

b. Sotatercept and Luspatercept: These are new class of drugs used subcutaneously to

prevent apoptosis of late stage erythroblasts, a major cause of anemia in thalassemia

syndrome. These drugs act as TGF activating ligand traps. These drugs are

considered to be a significant advance to treat this disease

c. Allogenenic bone marrow transplantation with HLA matched sibling or unrelated

donor is already an accepted mode of therapy of severe thalassemia and sickle cell

Page 284: Training Manual for Hemoglobinopathies and Hemophilia:

284

disease. Significant side effects of this modality of therapy must be kept in mind

before advising to go for such therapy.

d. Gene Therapy. This mode of therapy has advanced significantly now and already

patients with thalassemia and sickle cell anemia have been treated with significantly

improved results in USA.

References:

1. Vrettou C, Kakourou G, Mamas T, Traeger-Synodinos J. Prenatal and

preimplantation diagnosis of hemoglobinopathies.Int J Lab Hematol. 2018 ;40

(S1):74-82.

2. Colah RB, Gorakshakar AC, Nadkarni AH.Invasive& non-invasive approaches

for prenatal diagnosis of haemoglobinopathies: experiences from India.Indian J Med

Res. 2011 ;134:552-60.

3. D.J.Weatherall and J.B.Clegg ,R.GibbonsD.R.HiggsJ.M.Old Nancy F.OlivieriSwee

Lay Thein W.G.Wood. The Thalassaemia Syndromes Fourth edition (2001)

Blackwell publication Oxford.Reprinted 2010.

4. Taher AT, CappelliniMD.How I manage medical complications of β-thalassemia in

adults.Blood. 2018 ;132:1781-1791.

5. Cappellini MD, Cohen A, Porter J, ( Eds).Guidelines for the Management of

Transfusion Dependent Thalassaemia (TDT) [Internet]. 3rd edition,

2014.Thalassaemia International Federation; Nicosia (CY).

6. Serjeant GR & Serjeant BE,(eds) Sickle cell Disease. 3rd

Ed 2001. Oxford University

Press.

7. GreerJP , Arber DA, Glader B, List AF, Means Jr.RT, Paraskevas F, Rodgers GM,

Foerster J . (Eds). Wintrobe’s ClinicalHematology. 13 th Ed 2013.Wolter Kluwer,

USA.

8. Torres L, Conran N.Emerging pharmacotherapeutic approaches for

the management of sickle cell disease.ExpertOpinPharmacother. 2019 ;20:173-186.

9. Taher AT, Weatherall DJ, CappelliniMD.Thalassaemia.Lancet. 2018 ;391:155-167.

10. Algiraigri AH, Wright NAM, Paolucci EO, Kassam A.Hydroxyurea for lifelong

transfusion-dependent β-thalassemia: A meta-analysis.PediatrHematol Oncol. 2017

;34:435-448.

Page 285: Training Manual for Hemoglobinopathies and Hemophilia:

285

Registry for Hemoglobinopathies

Hemoglobinopathies are the most prevalent genetic diseases in India. The national prevention

program is running successfully, however as the patients grow older, their management gets

more complicated. Paper based clinical notes are being used and there are some electronic

data banks mainly for financial matters.

The importance of ongoing systematic collection, analysis, and interpretation of data on

Hemoglobinopathies affecting the population, closely integrated with timely dissemination of

these data to those responsible for prevention and control will be quite useful. The registry is

meant to have a reliable data bank for epidemiologic and clinical data for the patients

suffering from blood disorders. This registry will be a source for health system decision

makers. The registry should consist of epidemiologic data, clinical history and important

physical examination points, laboratory results, a list of classic complications, list of

medications.

The registry needs to focus on:

• Assessing the burden of disease

• Monitoring trends in health

• Identifying emerging risks

Each record should be updated at least one time in each season. Electronic Registry should be

an online data bank for patients, physicians and policy makers for better research and

development in the field of Hemoglobinopathies.

The policy for Hemoglobinopathies envisages a robust system of data collection to be entered

systematically in a registry. Data should be collected both of patients to assess treatment

needs for blood and medicines etc. and carriers.

This data collection also helps in collecting secondary data:

1. Disease severity, co-morbidities, and chronic disease complications of persons with

Hemoglobinopathies.

2. Disease and treatment-related infections.

3. Reproductive and pregnancy outcomes of hemoglobinopathy patient populations.

4. Mortality rates, including case fatality rates for Hemoglobinopathies and complications.

5. Health care utilization, costs of care, and the geographic variation of specific service etc.

The policy recommends setting up of a patient registry for thalassemia and sickle cell

disease to obtain information on the number of persons affected and the number of

carriers to estimate patients who require various services.

Page 286: Training Manual for Hemoglobinopathies and Hemophilia:

286

The data on carrier screening performed in different regions should be collated to

determine the burden of hemoglobinopathies.

Creation of Central Hematopoietic Stem Cell donor registry, to facilitate bone marrow

transplants in those patients lacking sibling donors.

Generated across a variety of sources, data collection in hemoglobinopathies will also

encourage efficient communication between doctors and patients, and increase the

overall quality of patient care. As a response tothe digitization

of healthcare information and the rise of value-based care, the health care department

has taken advantage of this big data and analytics to make strategic decisions.

Page 287: Training Manual for Hemoglobinopathies and Hemophilia:

287

Section B: Hemophilia

Section Diagnosis of Management of Haemophilia Page

no.

1 Introduction 288

2 Abbreviations 290

3 Definition of various Levels of Training 293

4 Diagnosis of Hemophilia and other Bleeding Disorders 296

i Setting up a coagulation laboratory 297

ii Laboratory approach to bleeding disorders 301

iii Preanalytical variables and Quality Assurance in a coagulation laboratory 306

iv Laboratory Diagnosis of hemophilia and other bleeding disorders 315

v Genetic Counselling for haemophilia 348

vi Genetic Diagnosis of haemophilia 353

vii Women with Bleeding Disorders: Approach and laboratory Diagnosis 362

5 Management of Haemophilia and other Bleeding Disorders 369

i Clinical manifestations and management of Hemophilia 370

ii Management of hemophilia with inhibitors 385

iii Implementation of prophylaxis in hemophilia patients in India 391

iv Surgery in haemophilia 397

v Cautions to be exercised while using newer products/novel therapeutic

approaches for haemophilia

407

vi A comprehensive model for conservative management in haemophilia 414

vii Psychosocial Care and Support: Training Curriculum for

Psychologists/Social Workers/Nurses working in Bleeding Disorders

422

Page 288: Training Manual for Hemoglobinopathies and Hemophilia:

288

Diagnosis and

Management of

Hemophilia

Page 289: Training Manual for Hemoglobinopathies and Hemophilia:

289

Introduction

Hemophilia A and hemophilia B are inherited single gene disorders with an incidence of 1

per 10,000-30000 male births respectively. Besides hemophilia, there is a wide range of other

bleeding disorders which include von Willebrand disease (VWD), rare coagulation factor

defects, platelet function disorders and defects in the fibrinolytic pathway. These disorders

may manifest as spontaneous or post- trauma hemorrhagic episodes in patients. The

diagnostic and therapeutic aspects are important as specific therapy is now available for many

of these disorders.

The epidemiological data on the number of haemophilia patients in India is lacking which is

important for any public health intervention. Translating the incidence of haemophilia in the

Western World, it is anticipated that in India there may be approximately around 80000-

100000 haemophilia cases; however the number of registered patients in the disease registry

of Hemophilia Federation of India (HFI), a non-governmental Organization is only around

19000. As far as the global rank is concerned, India reports the second highest number of

patients with haemophilia A and 9 per cent of the total number of patients with haemophilia

A in the World. Though this data is generated by a very well organized network of more than

90 local Hemophilia Chapters across the country, there is still a gross unawareness and

underdiagnosis of haemophilia in this country.

The bleeding manifestations in hemophilia generally depend on the residual factor levels.

These patients are arbitrarily classified as severe (factor levels <1%), moderate (factor levels

1-5%) and mild (factor levels 5-40%). The appropriate treatment haemophilia patients is

replacement therapy i.e plasma derived or recombinant factor VIII or factor IX, which is

highly resource intensive. Due to the exorbitant cost and scanty availability of these products

in India, it used to be out of reach for many of our hemophilia patients. But thanks to the

Humanitarian Aid Program of World Federation of Hemophilia; in many of the Government

Hospitals and other places, there is a continuous supply of factor products which caters to not

only for “on demand” therapy but several children are on prophylactic treatment in several

Centers at no cost . This has also enabled to put few inhibitor positive patients on Immune

tolerance induction protocol. The HFI is mainly responsible for the data exchange,

requisition, import and distribution of treatment products, patient counselling, training ,

education, link patients to needed services and advocate for the rights of patients with

hemophilia and other bleeding disorders. Besides this, several State Governments are also

providing factors free of cost to haemophilia patients.

Page 290: Training Manual for Hemoglobinopathies and Hemophilia:

290

While more and more treatment products are becoming freely available, the training,

education and diagnostic facilities pose a major problem in this area. There are very few

comprehensive diagnostic facilities for bleeding disorders in the country, besides the fact that

even the medical personnel treating the haemophilia patients is not thoroughly educated about

the optimum treatment protocols within the existing resources. The only way to overcome all

these limitations is to implement a National program for diagnosis, care, support and

preventive strategies.

Page 291: Training Manual for Hemoglobinopathies and Hemophilia:

291

Abbreviations

AAP assembly activating protein

AAV Adeno-associated Virus

AD Autosomal dominant

ADP Adenosine diphosphate

ANM Auxiliary Nurse Midwife

aPCC Activated Prothrombin Complex Concentrates

APTT Activated Partial Thromboplastin time

AR Autosomal recessive

ASAP As soon as possible

ASHA Accredited Social Health Activist

AV Adenovirus

bp Base pair

BSC Biological safety cabinet

BSL Biosafety level

BSS Bernard Soulier Syndrome

BT Bleeding time

BU Bethesda Units

CBC Complete Blood count

CD Cluster of differentiation

CFC Clotting factor concentrates

CHC Community Health Centre

CHO-KLAT Canadian Hemophilia Outcomes: Kids’ Life Assessment Tool

CNS Central nervous system

CS Clot solubility

CVS Chorionic villus sampling

D/W Distilled water

DEIC District Early Intervention Centre

DH District Hospital

DIC Disseminated intravascular coagulation

DNA Deoxyribonucleic acid

dNTP Deoxyribonucleotide triphosphate

DRVVT Dilute Russell’s Viper venom time

EACA Epsilon aminocaproic acid

ED Exposure days

EDTA Ethylenediaminetetraacetic acid

ELISA Enzyme-linked immunosorbent assay

EQAS External Quality Assurance Scheme

F(Roman no) Factor

FDP Fibrinogen degradation product

FEIBA Factor eight inhibitor bypass activity

Page 292: Training Manual for Hemoglobinopathies and Hemophilia:

292

FGFR-1 Fibroblast growth factor receptor 1

FISH Functional Independence Score in Hemophilia

GI Gastrointestinal

GP Glycoprotein

GT Glanzmanns’s Thrombasthenia

HA Haemophilia A

HAL Haemophilia Activities List

HB Haemophilia B

HCL Hydrochloric acid

HCV Hepatitis C virus

HIV Human immunodeficiency virus

HPV Human Pappiloma virus

HR Health-related

HRP horseradish peroxidise

HSPG Heparan sulphate proteoglycans

HSV Herpes Simplex virus

HTC Haemophilia training center

IC Intracranial

IEC Institutional Ethical Committee

INR International normalized ratio

IQC Internal Quality Control

ITI Immune Tolerance Induction

ITR Inverted terminal repeats

IU International units

IVF In vitro fertilization

kb Kilo base

LA Lupus anticoagulants

LT Laboratory Technologist

Mab Monoclonal antibodies

mM Milli Molar

MMR Measles, mumps, and rubella

MO Medical Officer

MRI Magnetic resonance imaging

MSK Musculoskeletal

NPP Normal pool plasma

NSAIDS Nonsteroidal anti-inflammatory drugs

NV Non variceal

OBS Owren’s barbiturate saline

OD Optical density

OPD 1,2-o-Phenylenediamine dihydrochloride

ORF Open reading frames

PBS Phosphate buffer saline

PCR Polymerase Chain Reaction

Page 293: Training Manual for Hemoglobinopathies and Hemophilia:

293

PF3 Platelet factor 3

PFD Platelet function disorders

PGD Preimplantation genetic diagnosis

PHC Primary Health Centre

PK Prekallikrein

PNPP p-nitrophenyl phosphate

PPP Platelet poor plasma

PRP Platelet rich plasma

PSS Principles of Psychosocial Support

PT Prothrombin time

PWH Persons with haemophilia

QA Quality assurance

QC Quality control

QoL Quality of life

RBSK Rashtriya Bal SwasthyaKaryakarm

rFVIIa Recombinant activated factor VII

RG1 Risk Group classification 1

RIPA Ristocetin induced platelet aggregation

ROTEM Rotational thromboelastometry

RT Reptilase Time

RVVT Russell’s Viper venom time

SDS sodium dodecyl sulphate

SOP Standard operating procedure

TEG Thromboelastography

TMB 3,3',5,5'-Tetramethylbenzidine

TOT Training Of Trainers

TT Thrombin time

UK United Kingdom

VWF Von Willebrand factor

WFH World federation of haemophilia

μL Microliter

Page 294: Training Manual for Hemoglobinopathies and Hemophilia:

294

Page 295: Training Manual for Hemoglobinopathies and Hemophilia:

295

Table 1. Definition of various Levels of Training

Training in comprehensive diagnosis and management of Haemophilia and other bleeding disorders at different levels of Health Care

System

Level Centers Designated

Centers

Infrastructure Personnel in

the Center

Personnel

to be

trained by

the Center

Tests Instruments Other

responsibilities

1 Primary Health

Center( PHC)

Community Health

Center (CHC)

- ASHA, ANM and

RBSK workers

- CBC ,

Peripheral

smear

Three-part automated

blood cell counters and

microscope

Refer to Level 2

2 Integrated centre

for

Hemoglobinopathy

and Hemophilia

District

Hospitals

Govt.

Medical

colleges.

- Medical officers

(MO)

Laboratory

technicians (LT),

Pathologists,

Counsellors

Obstetricians

Trainees of

trainers (TOT)

- CBC

Peripheral

smear

Screening

coagulation

tests

Mixing studies

Factor assays

Inhibitor

screening

Three-part automated

blood cell counters and

microscope

Centrifuges

Semiautomated

Coagulometers

Diagnosis and

management

Refer to Level 3

whereever

required

Page 296: Training Manual for Hemoglobinopathies and Hemophilia:

296

3 Center of

Excellence

1-2 per

State

Approximately

2500 square

feet with

lecture

theaters and

laboratory

space for

training

modules for

Level 1 and 2

participants

Social

counsellor, TOT

trainer,

Pathologist,

Obstetrician for

CVS, Clinical

Hematologist,

Molecular

Biologist

Designated

MOs and

staff at the

DH

District

Early

Intervention

Centre’s

(DEIC),

(Levels 1

and 2)

CBC

Peripheral

smear

Screening

coagulation

tests

Mixing studies

Factor assays

Inhibitor

screening

Nijmegen

modified

Bethesda assay

( NBA)

Platelet

function tests

Genetic

counselling and

Genetic

diagnosis

Three-part automated

blood cell counters

Microscope

Centrifuges

Semiautomated/ Fully

automated

Coagulometers

Platelet aggregometers

Flow Cytometers

PCR machine

Electrophoresis system

Gel documentation

system

Automated DNA

sequencer

Inverted microscope

for CVS dissection

Diagnosis and

management

Quality

Control: Both

External

Quality

Assessment (

EQAS) and

Internal

Quality Control

( IQC) to be

done regularly.

Concept of

Reagent Rental

with

maintenance of

equipment

could be used

which will

include EQAS.

4 National Training

Institutes*

ICMR-NIIH,

Mumbai,

AIIMS, New

Delhi

SGPGI,

Lucknow

CMC,

Vellore

- Personnel from

Levels 1-3

Level 3 Comprehensive

laboratory

diagnosis ,

Genetic

counselling and

Genetic

Diagnosis for

all bleeding

Three-part automated

blood cell counters

Microscope

Centrifuges

Semiautomated/ Fully

automated

Coagulometers

Platelet aggregometers

Quality Control

: The Center

should perform

both IQC and

EQA regularly.

Linkage Plan of

action between

Page 297: Training Manual for Hemoglobinopathies and Hemophilia:

297

PGIMER,

Chandigarh;

Calcutta

Medical

College,

Kolkata

disorders

Flow Cytometers

PCR machine

Electrophoresis system

Gel documentation

system

Automated DNA

sequencer

Inverted microscope

for CVS dissection

all the Levels

(1-4) will be

assured by the

National

Training

Institutes

*National Training Institutes and States to be covered by each Institute

1. ICMR-NIIH Mumbai: Maharastra, Gujrat, Goa, Daman & Diu, Dadra and Nagar Haveli, Karnataka

2. AIIMS, New Delhi: Delhi, Rajasthan, Madhya Pradesh, Chattisgarh

3. SGPGI, Lucknow: Uttar Pradesh, Bihar, Jharkhand

4. CMC, Vellore: Tamil Nadu, Kerala, Andra Pradesh, Telangana, Pondicherry, Andaman & Nicobar

5. PGIMER, Chandigarh: Punjab, Haryana, Himachal Pradesh, Uttarakhand, Jammu and Kashmir

6. Calcutta Medical College, Kolkata: West Bengal, Orrisa, Assam, Arunachal Pradesh, Meghalay, Sikkim, Manipur, Nagaland, Tripura

Page 298: Training Manual for Hemoglobinopathies and Hemophilia:

298

Diagnosis of Hemophilia and other

Bleeding Disorders

Page 299: Training Manual for Hemoglobinopathies and Hemophilia:

299

Setting up a Coagulation Laboratory with available health care

infrastructure

Coagulation laboratory is an essential component of good health care delivery for every

hospital.These laboratories come with different levels of sophistication. Four Levels of such

laboratories can be conceived within the existing Infrastructure.

Level1 :Basic coagulation laboratory which can perform screening coagulation tests

including platelet count and complete haemogramalong with mixing studies.. This should be

available at CHC/ Rural hospitals / Taluk hospitals. Depending on how PHCs are improving

their service, these types of laboratories can be integrated to PHCs also.

Level2 : These are the laboratories with basic coagulation work up facilitiesalong with factor

assayusing one stage APTT based technique andscreening tests for inhibitors. This kind of

laboratory should be available at District / Civil hospitals.

Level3 :This laboratory should have all the facilities of a Tier 2 coagulation laboratoryand in

addition should be able to perform platelet function tests, von-Willebrand factor (VWF) assay

and inhibitor levels by Bethesda assay. The laboratory should also do all rare coagulation

factor assays. It is desirable that this facility in collaboration with Department of

Gynaecology and Obstetrics should be able to develop prenatal chorionic villus sampling /

amniotic fluid aspiration facility and at least do basic molecular biology procedures like DNA

extraction and transfer these samples to toLevel 4 laboratories for molecular biology based

detection of haemophilia and other rare bleeding disorders. Level 3 laboratories at present

should be located in Medical colleges/ Centers of Excellence ( 1-2 per state).Level 3

laboratories should also look after quality control and training of manpower for Level 1 and

Level 2 laboratories.

Level 4: These are the State Apex laboratories and should have all the facilities of previous

laboratories i.e.Level1 -3 laboratories.This laboratory will be a reference laboratory for that

Page 300: Training Manual for Hemoglobinopathies and Hemophilia:

300

State and is also responsible for the Quality control and assurance in Level 3 Laboratories. In

addition, these laboratories should be a comprehensive diagnostic Center for all hemostatic

and thrombotic investigations.Some of the advanced diagnostic facilities like Flow cytometry

for platelet receptor analysis, a full pledged molecular diagnosis facility for all types of

bleeding disorders , bioinformatics facility and so on.ThisCenter is also responsible for all

types of trouble shooting, holding interactive sessions with its peers and other National level

laboratories. The major responsibility of this Center is training manpower and develop human

resources. Appropriate training modules need to be developed by these Centers.Level 4

laboratories should be located in all AIIMS / or in the top medical colleges of the states.These

laboratories should also be engaged in active research, at least in various pilot projects and

operational research with practical applicability.

Basic requirements of laboratories from Levels 1-4

Space :Additional space of at least 300 sq.ft , 500 sqft , 600 sq ft, 1200 sq ft is required for

Level 1 , Level 2 , Level 3 and Level 4 laboratories respectively.

Location :Level 1 and Level 2 laboratories can work as part of Hematology/ Pathology

laboratories. For Level 3 and Level4 , independent modules of laboratory servicesshould be

available.

Manpower: All the laboratories should be under the supervision of a Hematopathologist/

Hematologist/ Pathologist / Laboratory medicine expert / Doctor with at least two years of

experience of working in a coagulation laboratory as the case may be. However for Level 3

and Level 4 laboratories, the working experience of more than two years in a coagulation

laboratory should be preferred. Exceptions should be made for exceptional candidates.

For Level 1, provision for one Medical Laboratory Technician with experience in specific

area should be employed. A trained technician if already available should be transferred to

this area and a new personmeanwhile can be trained. It is desirable that all technicians should

at least be able to do screening coagulation tests (PT and APTT) and complete blood count

(CBC) so that in emergency situations, they can do the job.For. Level 2 and Level 3

laboratories, at least two Medical Laboratory Techniciansshould be recruited for each Centre.

They should have a genetic counsellor too. For Level 4 laboratories,at least three Medical

Laboratory Technicians , one genetic counselor along witha Scientist with higher

qualificationswith expertise and experience inadvanced techniques like molecular biology,

flow cytometry and so on. This person should be a postgraduate in Micobiology/Applied

Biology/Biochemistry or similar subjectsPh.D degree holders should be desirable.

Page 301: Training Manual for Hemoglobinopathies and Hemophilia:

301

Instrumentation:FromLevels 1-4, all laboratories need to have the following basic

equipments.

Semiautomated 2- channel/ 4 - channel coagulometer

Blood Cell counter

Good quality microscope.

4 o C Refrigerator and -40/-70

oC Freezers

Refrigerated centrifuge

Water bath/ Thermometer/ Stop watch

Automated pipettes

Computers with wifi connection

Level 3 laboratories, in addition need the following equipments

Nano drop spectrophotometer.

Platelet aggregometer.

ELISA Reader

Dissecting microscope.

Whole blood coagulation analyser ( ROTEM,

Thromboelastography)

Level 4 laboratories need all the instruments mentioned above for Tiers 1-3 , but in

addition need the following equipments

Flowcytometer

A good Molecular Biology Laboratory with automated DNA

Sequencer

Reagents and consummables

The reagents and other consummables may vary from laboratory to laboratory; however all

laboratories need the following

Reagents to run automated blood cell counters.

Leishman stain.

PT/APTT/TT Reagents

Control normal lasma/ Deficient plasma

Calibrators with known factor levels

Disposable plasticwares/ Glasswares

Level 3 and 4 laboratories should also get fluorescence labelled antibodies and Molecular

Biology reagents

Page 302: Training Manual for Hemoglobinopathies and Hemophilia:

302

Tests to be available at different Levels of healthcare facility

Level 1

Screening coagulation tests ie PT, APTT and TT and CBC

Mixing studies

Level 2

All tests done at Tier 1

Factor VIII and IX assays

Screening for inhibitor including progressive inhibitor

D- dimer/ FDP screening

Level 3

All tests done at Tier 1-2

Quantitative inhibitor assays (Bethesda assay)

Thromboelastography

Platelet aggregometry

VWF assay

CVS and DNA extraction

Level 4

As this is the Reference Laboratory in the State, these laboratories should have a

comprehensive diagnostic facilities for diagnosis of all bleeding and thrombotic disorders.

These Centers also should look into developing novel simpler techniques for diagnosis. The

laboratory should also mmake efforts for diagnostic investigations to monitor altered newer

products in the marketwhich pose additional challenges of investigation for these apex

laboratories.Every laboratory should have a standard operating procedure ( SOP) manual.

Page 303: Training Manual for Hemoglobinopathies and Hemophilia:

303

Basics of Laboratory approach to bleeding disorders

Bleeding disorders constitute an important group of disorders in Hematology. The duties of

Laboratory Hematologists include diagnosis of the bleeding disorders by doing relevant tests

and interpret them correctly. It is important to have relevant clinical information before

interpreting the results. From the history, it is important to differentiate hereditary disorders

from acquired. Hereditary disorders generally start early in age, recurrent in nature and may

have a positive family history. On the other hand, acquired disorders occur at any age and

may have an underlying predisposing cause. History of medication including history of

administration of replacement therapy, warfarin or heparin also helps in interpretation of

tests. Depending on the type of bleed, it is important to differentiate between platelet and

coagulation factor deficiency disorders. In general, platelet disorder related bleeds present as

petichealhemorrhage, mucosal bleeds, whereas coagulation factor disorder presents as deep

abdominal bleeds, muscle bleeds, epistaxis and ecchymotic patches.

Laboratory Approach

Since a large number of tests are available for diagnosis of the spectrum of bleeding

disorders, it is important to go in a stepwise approach. A short screening coagulation should

be the starting point. Depending on the abnormalities observed in the screening tests,

specialized diagnostic tests can be performed.

Figure 1. The Coagulation Cascade

Page 304: Training Manual for Hemoglobinopathies and Hemophilia:

304

RVVT: Russel Viper venom time ; PF3: platelet factor 3; CS: clot solubility

Table 2. Screening tests in a patient with bleeding disorder

Screening test Normal Defective

Platelet count (PC) 150-450/cumm

Bleeding time (BT) 2-5 minutes PC

Platelet function

Vascular function

Prothrombin time (PT) 12-14 seconds Extrinsic pathway

(FVII deficiency)

Common Pathway (FI, II, V,

X deficiency)

Activated Partial

Thromboplastin time(APTT)

35-45 seconds Intrinsic pathway ( FVIII, IX,

XI, XII deficiency)

Common pathway (FI, II, V,

X deficiency)

Clot stability (CS) Stable upto 24 hours F XIII deficiency

Bleeding time

Bleeding time (BT) is prolonged in thrombocytopenia and platelet function disorders(PFD).

If it is prolonged and platelet count is normal, diagnostic tests for PFD such as platelet

aggregation with ADP, Adrenalin, Arachidonic acid, Collagen and Ristocetin can be

performed for specific diagnosis.

Page 305: Training Manual for Hemoglobinopathies and Hemophilia:

305

Abnormalities in PT/APTT

The following combinations of abnormalities are possible for PT/APTT

Prolonged PT + Normal APTT

Normal PT + Prolonged APTT

Prolonged PT + Prolonged APTT

Isolated prolongation of PT may be due to hereditary FVII deficiency or acquired deficiency

of F VII as in mild liver disease or warfarin effect.

Isolated prolongation of APTT

This is seen in inherited deficiencies of intrinsic factors like FVIII, IX, XI and XII. In the

absence of facilities for factor assays, mixing studies of APTT with normal serum and

aluminium hydroxide adsorbed plasma can be performed. Normal serum contains factors IX,

X, XI and XII. Adsorbed plasma contains FV, FVIII, XI, XII. The interpretation is given as

follows:

Table 3. Approach to a patient with suspected coagulopathy

PT: Normal APTT ↑

Hereditary defect (VIII,IX,XI,XII)

Mixing studies (APTT)

Normal serum

corrected

Normal serum not

corrected

Normal serum not

corrected

Adsorbed plasma

not corrected

Adsorbed plasma

corrected

Adsorbed plasma not

corrected

Diagnosis FIX deficiency FVIII deficiency F XI,XII deficiency

VWD (↑BT)

Diagnostic test Factor assay

If prolonged APTT is accompanied by prolonged BT, VWD may be suspected . Sometimes,

however, in VWD variants, BT may not be prolonged .This can be confirmed using

Ristocetin induced platelet aggregation (RIPA), VWF Ag estimation ,VWF Ristocetin

cofactor assay and multimeric analysis. . Known abnormal plasma is also useful for

identification of factor defects by testing the ability of an unknown plasma to shorten the

prolonged clotting time of plasma with known defects. In this instance, patient should be

suffering from a severe defect (Factor level <1%). Acquired cause of isolated prolongation of

Page 306: Training Manual for Hemoglobinopathies and Hemophilia:

306

APTT includes presence of lupus anticoagulants, intrinsic factor inhibitors or heparin

therapy. Lupus anticoagulant (LA) can be confirmed by Kaolin clotting assay and dilute

Russell viper venom test (DRVVT). Factor inhibitors can be confirmed by screening tests of

APTT followed by inhibitor assay (Bethesda assay).

Prolongation of both PT and APTT

This indicates deficiency in common pathway of coagulation (FI, II, V and X) or multiple

coagulation factor defects which may be inherited or acquired. When history suggests

hereditary defect, mixing studies with normal serum or adsorbed plasma are helpful in

differential diagnosis as given below:

Table 4. Interpretation of mixing studies

Causes of prolonged PT and APTT

PT↑ APTT ↑

Hereditary defect (V,X,II,I)

Mixing studies (APTT / PT)

Normal serum

corrected

Normal serum not

corrected

Normal serum not

corrected

Adsorbed plasma not

corrected

Adsorbed plasma

corrected

Adsorbed plasma not

corrected

FX deficiency FV deficiency TT

If normal: FII

deficiency

If ↑:

Hypofibrinogenemia

Dysfibrinogenemia

Perform specific Factor assay

Acquired conditions like Disseminated intravascular coagulation (DIC), fibrinolysis, liver

disease, vitamin K deficiency may also lead to prolongation of both PT and APTT. DIC is

confirmed by estimation of D-Dimer and/or FDP. Fibrinolysis shows normal D-Dimer but

abnormal FDP. Liver disease can be confirmed by liver function tests (SGOT/SGPT).

Vitamin K deficiency can be confirmed by repetition of PT, APTT 48 to 72 hrs after giving

5mg of vitamin K intravenously.

Despite such extensive testing, normal screening tests can be obtained in the following

conditions.

Disorders with normal screening tests

•Mild clotting factor deficiency

•Simple purpura

Page 307: Training Manual for Hemoglobinopathies and Hemophilia:

307

•Senile purpura

•Henoch Schonlein purpura

•Scurvy

•Hereditary hemorrhagictelengiectasia

Normal Control

These should be taken from healthy people not known to be suffering from any disease and

not from so called ‘normal’ patients (i.e. patients whose disease does not apparently seem to

affect coagulation system). Samples should not be collected from individuals under certain

conditions which might stimulate adrenoreceptors (e.g vigorous exercise such as running up

stairs ,donating blood transfusions etc.), which raise factor VIII in a few minutes or evoke

the ‘acute phase reaction’ raising fibrinogen, Factors V and VIII .Samples should not be

collected when a person has infection or from pregnant people.The wide range of normal

factor levels severely limits the usefulness of single random sample as control. Use of pooled

plasma from a number of healthy donors eliminates this variation to a considerable extent.

Abnormal Control

These are useful in the preparation of certain reagents. For instance the sensitivity of

phospholipid platelet substitute can be judged from the APTT performed on plasma from a

haemophilic with a mild defect(Factor VIII around 20%).

Page 308: Training Manual for Hemoglobinopathies and Hemophilia:

308

Preanalytical variables and Quality Assurance in a coagulation laboratory

The term ‘pre-analytical phase’ describes all actions and aspects of the medical laboratory

diagnostic procedure that occurs prior to the analytical phase.

These are factors that may influence the state of the sample and is the most vulnerable

part of the total testing process.

It encompasses all procedures, starting with the formulation of the medical question,

and includes patient preparation, sample collection, handling, transportation,

processing, and storage until time of analysis.

Biggest source of laboratory error; far exceeds analytical error.

Upto 70% of the errors take place in the pre-analytic phase.

Figure 2. Errors in different phases of testing

70-80% of clinical decisions are based on laboratory results.

Some of the common pre-analytical variables

False identification of the samples

Use of inadequate devices or needles

Incorrect order of draw

Prolonged tourniquet placing

Unsuccessful attempts to locate the vein

Incorrect use of additive tubes

Collection of unsuitable samples (e.g. contaminated, haemolysed) or quantity (e.g.

insufficient amount of blood or inappropriate blood-to-anticoagulant ratio)

Inappropriate mixing of a sample

Page 309: Training Manual for Hemoglobinopathies and Hemophilia:

309

Following are the areas where ‘Pre- analytical variations and errors’ can occur, which may be

addressed by following the suggested recommendations

Clinical history and physical examination

Bleeding history, bleeding score and family history

Drugs like aspirin or other antiplatelet drugs, NSAIDs, etc. Drug-induced effects on

platelet function should be considered when interpreting results.

Hormonal contraceptives

Bleeding history in relation to surgical, dental and obstetrical procedures.

Blood Group : Patients with O blood group are known to have lower levels of VWF.

Pregnancy status

Before sample collection

High fat meals interfere with light transmission aggregometry; 8- 12 hours fasting

samples are preferable

Smoking , consumption of alcohol, caffeine,physical activity , stress

During sample collection

Labeling must be done, before venipuncture, in the presence of the patient.

Blood sample should be collected directly from a peripheral vein (antecubital vein).

Tourniquet should not be used for over one minute.

The guage of the needle should preferably be between 19 and 22.

Blood samples for Haemostasis testing should not be obtained from an existing vascular

access device such as an intravenous (IV) line or a central line, which contain heparin.

Vacuum aspiration of blood into collection tubes may have adverse effect on platelet

function testing.

A syringe system permitting slow manual drawing of blood may be preferred.

Sample tubes and anticoagulants

Use siliconized glass or plastic (polypropylene) tubes.

Blood samples should be drawn into 105–109 mmol/L sodium citrate, buffered

anticoagulant.

The pH of the anticoagulated plasma should be between 7.3 and 7.45.

Collect venipuncture directly from a peripheral vein (antecubital vein).

The order of drawing blood during phlebotomy should be blood culture/sterile tubes,

then coagulation tubes, then plain tubes/gel tubes, then tubes containing additives.

Page 310: Training Manual for Hemoglobinopathies and Hemophilia:

310

Collect a discard tube when citrated plasma is obtained using butterfly needles.

The ratio of sodium citrate to whole blood must be 1:9.

In case of high haematocrit, the citrate volume should be adjusted maintain the citrate-

calcium ratio, and avoid plasma dilution with excess citrate.

Needed citrate volume (C, mL) may be calculated according to CLSI formula

C=(1.85x10-3) x(100-Hct)xV (where, V = original tube volume).

Figure 3. Citrate concentration in relation to hematocrit

Sample processing

Blood should be adequately, gently and promptly mixed by 4 to 6 complete end-over-end

inversions of the tubes in order to ensure even mixing of anticoagulant with blood.

Vigorous shaking, vortexing or agitation of blood samples should be avoided.

In case of high Haemocrit, Citrate volume adjustment must be done.

Examine blood sample tubes for presence of clots, precipitates or haemolysis, before

processing.

Blood sample transportation

Before transport, sample tubes should be examined for identification, safety conditions

and stability.

For local transportation, transport samples at ambient temperature (15–25°C), so as to

reach the testing laboratory within an hour of collection.

For long distance transportation, plasma should be separated, distributed in small

aliquots, and snap frozen in liquid nitrogen. The samples should be transported in frozen

condition.

Page 311: Training Manual for Hemoglobinopathies and Hemophilia:

311

Blood samples must remain capped for transport.

Sample rejection

Transported samples received in appropriate temperature conditions.

Inappropriate collection tubes and anti-coagulant.

Expired anti-coagulant tubes.

Patient identification ambiguity or lack of identification.

Inappropriate blood volume in relation to anti-coagulant.

Haemolysed samples.

Clot in the blood / Plasma sample.

Centrifugation

For obtaining platelet-rich plasma (PRP) for platelet function analysis, centrifugation

should be performed at 200–250 g for 10 min without application of a rotor brake.

Refrigerated centrifuge at 4°C should be used for processing samples for coagulation

assays.

Centrifuge the sample tube for coagulation testing at 1500g, 15 min.

The preparation of Platelet Poor Plasma (PPP) requires double centrifugation to obtain a

residual platelet count lower than 10x109/L.

Following initial centrifugation, carefully transfer the plasma to a non- activating plastic

centrifuge tube using an auto- pipette, and then centrifuged again for about 15 min.

There should be no vibrations or imbalance in the centrifuge.

Sample storage

Store samples at room temperature (15–25°C), to be processed and analyzed within 2- 4

hours of collection.

Perform whole blood assays < 4 hr after blood sampling and centrifugation < 1 hr.

Store PPP at −80 °C for long term storage (18- 24 months).

If whole blood sample is centrifuged within 1 hr of collection, the plasma can be left on

top of the cells at room temperature up to 4 hrs prior to assaying.

Time from sampling to analysis depends on analyte

Samples for PT/INR have longer stability (24 hrs) at room temperature (15–25°C).

Samples for APTT should be performed using fresh plasma < 4 hrs (<1 hr in patients

treated with unfractionated heparin).

For platelet function assays, samples should rest at room temperature for at least 15 min

before analysis. Testing should be completed within 3– 4 hrs of collection.

Page 312: Training Manual for Hemoglobinopathies and Hemophilia:

312

Factors V and VIII assays should be performed within 3 hrs of collection.

Protein S shows significant loss of activity at 8 hrs.

VWF appears to be stable at 15–25°C for 48 h.

Freezing and thawing

Centrifuge samples that cannot be tested within 4 hrs and freeze the plasma aliquot.

Use rapid freezing technique (liquid nitrogen).

Store samples at −70 °C (or below) rather than −20 °C.

Plasma samples frozen at minus 20 °C remain stable for 2 weeks.

Plasma frozen at minus 80 °C remains stable for 6 – 24 months.

Do not re-freeze samples (prepare sufficient number of aliquots).

Thaw samples rapidly at 37 °C (to prevent denaturing fibrinogen) at least 5 min in a

water bath at 37 °C and not at room temperature, or work bench or in a microwave oven.

Test immediately.

After thawing, mix the sample gently to re-suspend any cryoprecipitate. Do not vortex or

shake.

In a coagulation laboratory, it is essential to ensure that the right test is carried out with the

right blood sample; after which the correct results are issued to the specific patient without

any undue delay.

Quality control and assurance

Quality assurance (QA) is defined as the overall programme for achieving these objectives

whereas Quality control (QC) is defined as measures that needs to be included for each assay

performed to ensure that the test is working properly. The term ‘QC refers to the statistical

quality control that is commonly used in laboratories to monitor the routine performance of

testing processes, detect possible errors and correct problems before test results are reported.’

Quality Assurance Quality Control

An overall management plan to guarantee

the integrity of the data

A series of analytical measurements used to

assess the quality of the analytical data

Page 313: Training Manual for Hemoglobinopathies and Hemophilia:

313

Figure 4. The Quality Assurance Cycle

Components of Quality Control

The main aim of internal quality control procedures and external quality assessment programs

is to evaluate and improve quality in clinical laboratories.

Internal Quality control (IQC)

Monitoring all the laboratory tests done and includes measurements of specially

prepared materials and repeated measurements on routine specimens, along with daily

statistical analysis of the data.

It is primarily a demonstration of precision

It ensures continual evaluation of the reliability of the daily work of the laboratory

with validation of tests before reports are issued

Quality control for coagulation tests

Page 314: Training Manual for Hemoglobinopathies and Hemophilia:

314

Preparation and selection of reagents

Normal pooled plasma (NPP) preparation

For all coagulation factor assays, normal pooled plasma or unicalibrator with a known factor

level should be used as a control. Fresh control plasma from one healthy individual should

never be used as a control for factor assays.

Factor deficient plasma

Either “in house” deficient plasma or commercial deficient plasma may be used for factor

assays.

Reagent selection for PT and APTT

Calibration of centrifuge and pipettes

Page 315: Training Manual for Hemoglobinopathies and Hemophilia:

315

Schedule for quality control procedures

1. Calibration with reference standards

Instruments, pipettes After every 6 months or more frequently if

control chart or EQA indicates bias or

fluctuation in results

After any repair/ service

Others: thermometers, scales,

etc

Yearly

2. Control chart with control reagents

Every single day and with each batch of samples

For 2- 3 patient’s samples, duplicate tests needs to be done if there are

discrepancies

3. Patient’s test results analysis

Check previous tests and if changes in clinical state and analyse before issuing

report

4. EQAS performance

Monthly assessment

External Quality Control (EQA)

Evaluation by an outside agency of the laboratory and methods. This can be organized

nationally, regionally or internationally.

EQA ultimate and overarching goal is to improve the quality of laboratory services, along

with many other benefits as follows:

1. To enhance patient care and safety through improved laboratory practice

2. To characterize test bias and imprecision across multiple methods

3. To correlate specific method variables with bias and imprecision

4. To identify interfering substances and quantify their effects across multiple methods

5. To provide clinical laboratories with reliable information for replacing unsatisfactory

methodologies

6. To identify clinical laboratories that are at risk for poor performance

Page 316: Training Manual for Hemoglobinopathies and Hemophilia:

316

7. To satisfy accreditation and regulatory requirements

Source: Modified from Cunningham MT, Brandt JT, Chandler WL, et al. Quality assurance

in hemostasis: the perspective from the College of American Pathologists proficiency testing

program. Semin ThrombHemost2007;33:250–258.

Few of the Quality control exercises in Haemotology are as follows

CMC EQAS

UK NEQAS

EQAP

Suggested Reading

Westgard JO. Internal quality control: planning and implementation strategies. Ann Clin

Biochem 2003;40: 593–611.

Raiyani Ankit. Internal Quality control in coagulation lab. Sahayadri Speciality Hospital.

https://www.slideshare.net/adraiyani/internal-quality-control-iqc-in-coagulation-lab

Tietz. Fundamentals of Clinical Chemistry and Molecular Diagnostics - E-Book by Carl

A. Burtis, David E. Bruns. Chapter: Quality Management by George G klee and James

Westgard.

Cristiane F. Oliveira; Talma R. L. Fernandes. Analysis of the pre-analytical phase in a

private pathology laboratory of Maringá city-PR, Brazil. J Bras Patol Med Lab 2016; 52:.

78-83.

WHO Guidelines on Drawing Blood: Best Practices in Phlebotomy. Geneva; 2010.

http://www.euro.who.int/__data/assets/pdf_file/0005/268790/WHOguidelines-on

drawing-blood-best-practices-in-phlebotomy-Eng.pdf?ua=1.

Mackie I, Cooper P, Lawrie A, Kitchen S, Gray E, Laffan M, British Committee for

Standards in H. Guidelines on the laboratory aspects of assays used in haemostasis and

thrombosis. Int J Lab Hematol. 2013;35:1–13.

Harrison P, Mackie I, Mumford A, Briggs C, Liesner R, Winter M, Machin S. British

committee for standards in H. Guidelines for the laboratory investigation of heritable

disorders of platelet function. Br J Haematol. 2011;155:30–44.

Bonhomme F, Ajzenberg N, Schved JF, Molliex S, Samama CM, French A. Intensive

care committee on evaluation of routine preoperative T, French society of a, intensive C.

Pre-interventional haemostatic assessment: guidelines from the French society of

anaesthesia and intensive care. Eur J Anaesthesiol. 2013;30:142–62.

Magnette A, Chatelain M, Chatelain B, Ten Cate H, Mullier F. Pre-analytical issues in

the haemostasis laboratory: guidance for the clinical laboratories. Thromb J. 2016 Dec

12;14:49.

Favaloro EJ, Lippi G, Adcock DM. Preanalytical and PostAnalytical variables: The

leading cause of diagnostic error in hemostasis? Semin. Thromb. Hemost. 2008;34:612-

634.

CLSI. Collection, Transport, and Processing of Blood Specimens for Testing Plasma-

Based Coagulation Assays and Molecular Hemostasis Assays; Approved Guideline-

Fifth Edition. CLSI document H21-A5. Wayne, PA: Clinical and Laboratory Standards

Institute; 2008.

Page 317: Training Manual for Hemoglobinopathies and Hemophilia:

317

DetailedLaboratory Diagnosis of Hemophilia and other Bleeding Disorders

Defects of the coagulation, platelet, endothelial or fibrinolytic systems may lead to bleeding

and thrombotic abnormalities. While acquired defects may involve multiple systems;

congenital defects are usually due to an abnormality of a single factor. For providing an

accurate and specific diagnosis application of specific laboratory tests along with an

appropriate history and clinical background.

A highly regulated cascade of reactions involving platelets and coagulation proteins result in

the formation of a blood clot following an injury. This is reversible and self-limited. Initially

platelets adhere to collagen through specific receptors in the presence of von Willebrand

factor (VWF) upon exposure of subendothelium resulting in subsequent activation and

aggregation of platelets which forms a “platelet plug” at the injury site. Subsequently the

coagulation cascade gets activated resulting in a stable fibrin clot. Specific inhibitors then

inhibits each of the coagulation factors preventing clot extension beyond the site of injury.

The final step involved the fibrinolytic step which involves the lysis of fibrin in the blood

clot; thus restoring the blood flow.

Three important parameters are necessary for the diagnosis of a bleeding patient: a) family

history b) clinical history and c) laboratory test results.

Family history & pedigree

This should mainly consist of details of the affected members in the family like gender of the

affected individuals, detailed history of bleeding during surgery or bleeding in any of the

family members, death due to unknown cause along with other details relevant to each

individual family, history of consanguinity among parents. An extended family pedigree

should also be drawn with details of both dead and live members in the family.

Clinical history

This includes type of bleeding (deep tissue or superficial bleeding, joint bleed), spontaneous

or traumatic, menorrhagia or excessive bleeding during dental extraction/ surgery. Other

details like number of bleeding episodes and factor infusion annually, sites of bleeding,

response to treatment should also be noted in the clinical proforma.

Page 318: Training Manual for Hemoglobinopathies and Hemophilia:

318

A detailed family history including family pedigree along with the clinical details will give a

hint of diagnosis in many of the cases.

Laboratory tests

Laboratory tests include both screening and confirmatory tests to detect the deficiency or

abnormality of each of these factors.Screening tests include peripheral smear, platelet count,

prothrombin time (PT), activated partial thromboplastin time (APTT) and thrombin time

(TT). The screening test results gives a clue as to which confirmatory or specific tests should

be chosen and done; making these tests highly cost effective. Confirmatory tests include

platelet aggregation tests, factor assays, platelet receptor studies, von Willebrand factor

(VWF) assay, VWF multimer studies, VWF-FVIII binding assay, VWF-ristocetin cofactor

assay, VWF-collagen binding assay, Nijmegen modified Bethesda assay for inhibitors. For

the diagnosis of rare platelet function defects, there are another battery of other tests which

are performed in specific cases.

Coagulation Screening Tests

A broad overview of hemostatic defects is provided by the coagulation screening tests which

further provide clues as to which specific tests need to be performed.

a. Prothrombin time (PT)

Principle

The extrinsic pathway of coagulation is measured by this test. Activation of extrinsic clotting

factors is done by adding tissue factor (extracted from brain tissue or synthetic),

phospholipids and calcium ions to plasma resulting in thrombin generation and fibrin clot

formation. The test is sensitive to vitamin K dependent factors i.e. factors II, VII, V, X as

well as fibrinogen. Primarily this test is used for monitoring of anticoagulant therapy.

Requirements

Control and patient citrated plasma

Thromboplastin reagent

Page 319: Training Manual for Hemoglobinopathies and Hemophilia:

319

Procedure

Test plasma (50 μL)

Incubate for 1 minute at 37°C

Thromboplastin reagent (100 μL)

Note the clotting time

Normal Range: 12 - 14 seconds

Interpretation

A prolonged PT may be observed in the following conditions:

a. Congenital factor VII deficiency in cases of isolated PT prolongation in the absence

of other acquired causes leads to the diagnosis of

Congenital deficiency of factors II, V, VII, X

Severe fibrinogen deficiency

Inherited or acquired defects due to consumption (e.g. DIC)

Liver disease (vitamin K deficiency or lack of synthesis)

Dilution (massive transfusion)

b. In the presence of lupus anticoagulant

c. In the presence of specific inhibitors to coagulation factors

d. In the presence of high levels of heparin

e. Very low levels of fibrinogen

f. Patients on warfarin

b.Prothrombin time – International normalized ratio (PT-INR) determination

The INR provides a standardized scale

For monitoring patients who are under stable oral anticoagulant therapy, the INR provides a

standardized scale. The International Sensitivity Index (ISI) is calculated according to the

following formula:

INR = RISI

Where R = Patient PT

Control PT

An ISI value is assigned by calibration against an International reference preparation

according to the WHO recommendation for every lot of thromboplastin reagent. For

mechanical and photo-optical coagulation instruments, Lot-specific ISI values are provided.

Page 320: Training Manual for Hemoglobinopathies and Hemophilia:

320

The recommended therapeutic range is between an INR of 2.0 and 4.5 for patients on oral

anticoagulant therapy.

c. Activated partial thromboplastin time (APTT)

Principle

In this test, the intrinsic coagulation pathway is triggered by an activator such as kaolin which

activates the contact factors. The contact factors cause a cascade of enzyme reactions in the

presence of calcium ions and phospholipids (partial thromboplastin) resulting in the

generation of thrombin and fibrin clot formation. An abnormal APTT usually indicates

deficiency of factors VIII or IX and occasionally of XI or XII; if prothrombin time is normal.

Requirements

Control and patient citrated plasma

APTT reagent

0.025M Calcium Chloride (CaCl2)

Procedure

Test plasma (50 μL) + APTT reagent (50 μL)

Mix and incubate at 37°C for 3 or 5 minutes

(as per the instructions)

Pre-warmed 0.025M CaCl2 (50 μL)

Note the clotting time

Normal range: 30 – 35 seconds

Interpretation

A prolonged APTT may be observed in the following conditions:

a. Isolated APTT prolongation in the absence of other acquired causes provides a diagnosis

of hemophilia A, hemophilia B or rarely of factor XI, factor XII or contact factor

deficiency.

b. Congenital deficiency of factors II, V, VIII, IX, X, XI, XII, prekallikrein (PK), high

molecular weight kininogen (HMWK) or fibrinogen

c. It may be acquired e.g. DIC, liver disease

d. Vitamin K deficiency or decreased synthesis, dilution (massive transfusion), or

anticoagulation (coumarin derivatives)

e. Patient on Heparin therapy

f. Presence of specific inhibitors

Page 321: Training Manual for Hemoglobinopathies and Hemophilia:

321

g. Presence of lupus anticoagulant

Differential Sensitivity of APTT reagents

APTT reagents are composed of synthetic phospholipids or rabbit brain phospholipid extracts

along with negatively charged activators like kaolin, ellagic acid, silica or celite. The in vitro

clot formation is triggered by the activation of factor XII when an APTT reagent is added to

plasma. The APTT reagent is responsive to deficiencies in the intrinsic and/or common

pathway. Generally factor deficiencies below 30IU/dL are detected by the APTT reagents.

APTT reagents are also used for monitoring the anticoagulant effect of unfractionated

heparin. For monitoring heparin therapy, laboratories generally establish a reagent-specific

APTT therapeutic range.For LA screening, one should use two APTT reagents i.e. one with

low phospholipid content and one with high phospholipid content. If the prolonged APTT

observed by using low phospholipid content is corrected by using high phospholipid reagent;

it means there is the presence of LA. This however needs to be confirmed with specific

assays. Different sensitivities have been seen in different APTT reagents not only for

different coagulation factors or in different clinical settings, but also to different levels of

factors

Note: Though PT and APTT values generally correspond to factor levels, this may not hold

true in all cases. High levels of one clotting factor can compensate for the lower levels of

other factor. Another important point to note is that in the presence of any history of bleeding,

it is justified to go ahead and do specific factor assays.

d. Mixing studies for prolonged PT and APTT

For plasma samples found to have prolonged screening tests (i.e. PT/ APTT), further

investigations are required to detect the specific factor deficiency by performing mixing tests.

However, it is important to rule out the presence of an inhibitor.

Principle

If prolonged PT and APTT does not get corrected or if there is <50% correction after mixing

patients plasma with NPP, the presence of an inhibitor is indicated. Factor deficiency will be

indicated by similar tests performed with specific factor deficient plasma samples will

indicate factor deficiency, if there is no correction with that specific deficient plasma.

Requirements

Citrated plasma of control and patient

Page 322: Training Manual for Hemoglobinopathies and Hemophilia:

322

Factor deficient plasma

APTT reagent

APTT mixing study results for detection of specific factor deficiency:

Reagent APTT (seconds)

NPP 29

Patient 82

Patient + NPP 30

Patient+FVIII deficient plasma 83

Patient +FIX deficient plasma 36

Diagnosis: FVIII deficiency

e. APTT screening test for contact factor deficiencies

Principle

When a sample with markedly prolonged APTT, incubated with APTT reagent for a longer

time i.e. at least 3 times the original incubation time, results in normalized APTT;

deficiencies of contact factors i.e. Prekallekrein (PK) and High molecular weight kininogen

(HMWK) is suggested.

Requirements

Citrated plasma of control and patient.

APTT reagent

0.025M Calcium Chloride (CaCl2)

Procedure

Test plasma (50 μL) + APTT reagent (50 μL)

Mix and incubate at 37°C for 15 minutes

(in place of 5 minutes)

Pre-warmed 0.025M CaCl2 (50 μL)

Note the clotting time

Interpretation

Contact factor deficiency is suggestive if the prolonged APTT gets normalized with

prolonged incubation. Deficiency of contact factors is not related to bleeding; however it is

still necessary to explain the prolonged APTT, especially during pre-surgical evaluation.

Page 323: Training Manual for Hemoglobinopathies and Hemophilia:

323

f. Thrombin time (TT)

Principle

The formation of a fibrin clot in plasma is measured by thrombin time test by the action of

thrombin on fibrinogen. Abnormal TT is observed in cases of fibrinogen deficiency,

abnormal fibrinogen or inhibition of conversion of fibrinogen to fibrin by heparin or fibrin

degeneration products. It is also seen in liver failure and intravascular clotting.

Requirements

Citrated plasma of control and patient

Thrombin reagent

Procedure

Test plasma (100 μL)

Incubate at 37°C for 1 minute

Thrombin reagent (100 μL) (3NIH U/mL)

Note the clotting time

Normal Range: 12-14 seconds

g. Thrombin time (TT) with protamine sulphate neutralization for the detection of

presence of heparin

In the presence of unfractionated heparin and sometimes even in the presence of low

molecular weight heparin, TT is prolonged. This can be checked by adding protamine

sulphate to the TT reagent. Protamine sulphate neutralizes heparin in the plasma sample. A

working solution of 40 mg% solution is prepared in normal saline. One part of this solution is

mixed with 9 parts of TT reagent and the TT test is repeated as mentioned above. If the

thrombin time gets normalized after this addition, it may be confirmed that the prolonged

thrombin time is due to the presence of heparin in the sample.

Page 324: Training Manual for Hemoglobinopathies and Hemophilia:

324

h. Reptilase Time (RT)

Principle

Reptilase is an enzyme which is extracted from the venom of the snake named Bothropsatrox.

This enzyme is not affected by the presence of heparin; thus used in a reagent and the test

performed whenever a patient is on heparin. Reptilase reagent is also more sensitive to

abnormal fibrinogen and is thus used as a test for dysfibrinogenemia in some of the

laboratories. The RT has been found much more prolonged than TT in case of

dysfibrinogenemia.

Requirements

Control and patient citrated plasma

Reptilase time reagent: 1.7 mg/mL stock solution of the Reptilase reagent is prepared in

Owren’s buffer and is preserved in -700C in small aliquots. Working solution is prepared

by diluting the stock 1:10 times in Owren’s buffer.

Note: Avoid any contact with the reagent; masks and gloves should be worn and care

should be taken not to inhale the reagent.

Procedure

Test plasma (150 μL)

Incubate at 37°C for 1 min

Reptilase time reagent (50 μL)

Note the clotting time

Normal Range: 15-18 seconds.

Table 5. Thrombin time and Reptilase time Interpretation

Reptilase time (RT) Thrombin time (TT) Interpretation

Prolonged Prolonged Afibrinogenemia,

Hypofibrinogenemia,

DIC

More prolonged than TT Prolonged Dysfibrinogenemia

Normal Prolonged Heparin

Note:

i. For all of the above tests, volume of all the reagents mentioned is for the semi-

automated coagulometers.

ii. The normal range may vary from laboratory to laboratory.

Page 325: Training Manual for Hemoglobinopathies and Hemophilia:

325

Table 6. Interpretation of Screening coagulation test results

Prolonged Factor deficiency Gene defect Acquired causes

PT FVII F7 Liver disease, Oral

anticoagulants, DIC, Sepsis

APTT FVIII, FIX, FXI, FXII &

Contact factors

F8, F9, F11 ,F12,

& contact factor

genes

Liver disease, Oral

anticoagulants, Sepsis, DIC,

Heparin

TT or RT Fibrinogen FGA, FGB, FGG Sepsis, Liver disease, DIC

PT and

APTT

FV, FX, Combined

deficiency of FV

&FVIII, MCFD

F5,F10,LMAN1,

MCFD2

Sepsis, Liver disease, DIC

PT,APTT

and TT

Fibrinogen, FII FGA, FGB, FGG,

F2

Sepsis, Liver disease, DIC

4. Fibrinogen estimation

Fibrinogen disorders are be of 3 types:

a) Afibrinogenemia: Grossly decreased fibrinogen levels is seen; with very low and

undetectable plasma fibrinogen. Prolonged PT, APTT and TT along with abnormal

platelet function is seen

b) Hypofibrinogenemia: Mild to moderate decreased fibrinogen levels

c) Dysfibrinogenaemia: Abnormal fibrinogen function.

The ratio of fibrinogen levels measured by clot based and immunological assays is

disproportionate. Some patients have both reduced levels of fibrinogen and abnormal

function: hypodysfibrinogenaemia.

a. Modified Clauss assay

Principle: The clotting time of diluted plasma has a direct bearing on plasma fibrinogen

levels in the presence of excess of thrombin. A graph prepared by plotting a series of

dilutions of a reference plasma sample of known fibrinogen concentrations is used to

compare the test result.

Requirements:

Control and patient citrated plasma

Glyoxaline buffer, Imidazole buffer or Owren’s buffer (pH 7.35)

Page 326: Training Manual for Hemoglobinopathies and Hemophilia:

326

Thrombin reagent (100 NIH Units/mL)

Procedure:

Prepare 1:5, 1:10, 1:20 dilutions of normal plasma and patient’s plasma in Owren’s buffer

Take 50 µL in the cuvette of the coagulometer

Incubate at 37°C for 60 seconds

Add 50 μL of pre-warmed thrombin reagent

Note the clotting time

Plot in a log-log graph paper with clotting time in Y-axis and fibrinogen concentration in Y-

axis.

Normal range: 150- 350 mg/dL.

b. ELISA - Enzyme linked immunosorbent assay

Enzyme-linked immunosorbent assay (ELISA) are also used for measuring Fibrinogen using

human fibrinogen ELISA kit. Fibrinogen concentration and not functional activity are

measured by immunological assays. They hold value in the investigation of congenital

dysfibrinogenemia where there is a discrepancy between antigen level and functional activity.

Diagnosis of afibrinogenemia is given when both fibrinogen antigen and activity level is less

than 50 mg/dL. In case of hypofibrinogenemia, fibrinogen level ranges between 50 to 150

mg/dL. Normal or abnormal screening coagulation tests with normal fibrinogen antigen

levels i.e. 150 to 350 mg/dL is seen in dysfibrinogenemia cases; however the functional

activity is abnormal.

Different ELISA kits with different working principles are available commercially for

estimating fibrinogen antigen.

5. Factor XIII (FXIII) estimation

The screening tests for FXIII i.e. clot solubility assays, FXIII activity tests and quantitative

FXIII antigen tests for total FXIII and for FXIII-A and FXIII-B subunits are the available

methods for FXIII deficiency. The most widely used test in routine hemostasis laboratories

for many years is the clot solubility test which is an old and inexpensive technique. But only

severe FXIII deficiency is detected in this test. For clinical use, the quantitative assays are

important and thus both antigenic and functional assays are performed in most of the

laboratories now.

Page 327: Training Manual for Hemoglobinopathies and Hemophilia:

327

a. FXIII screening techniques

i) Urea clot lysis technique (only severe F XIII deficiency detection)

Principle

Factor XIII (FXIII) is a transglutaminase enzyme serving as the fibrin-stabilizing factor. It

cross links fibrin strands within the clot. During the clotting of blood, FXIII is activated; for

which calcium ions and thrombin are necessary. The process of stabilization of fibrin clot by

activated FXIII is known as transamidation. This stable fibrin clot is insoluble in 5M Urea

solution, which is not the case in the clot formed from FXIII deficient plasma; these clots are

soluble within 1- 24 hours in 5M urea.

Requirements

Control and patient citrated plasma

0.025M CaCl2

5M Urea

Procedure

Add patient plasma and control plasma (200 μL) in 2 separate glass test tube

Add pre-warmed 0.025M CaCl2 solution (200 μL)

Mix & incubate at 37°C (water bath)

for 30 minutes

Add 3 mL of 5M Urea solution

Incubate at room temperature (RT) overnight

Check for clot lysis

Interpretation

Observation Interpretation

Fibrin clot stable FXIII 2-100%

Fibrin clot lysed Severe FXIII deficiency (FXIII<2% )

ii) Acetic acid clot lysis method (mild to moderate FXIII deficiency detection)

Principle

Page 328: Training Manual for Hemoglobinopathies and Hemophilia:

328

In the presence of thrombin, a stable fibrin clot in case of normal plasma is insoluble in 2%

acetic acid solution, whereas clots formed from FXIII deficient plasma (<1-18%) are soluble

within 24 hours

Requirements

Control and patient citrated plasma

Thrombin reagent, bovine thrombin (10 NIH Units/mL)

2% acetic acid solution

5M Urea

Procedure

Tube 1 Tube 2

Patient plasma/Control plasma (200 μL)

Pre-warmed thrombin solution (10 NIH

Units/mL) (200 μL)

Pre-warmed 0.025M CaCl2 solution

(200 μL)

Mix & incubate at 37°C (water bath) for 30 minutes

2% Acetic acid solution (3mL) 5M Urea solution (3mL)

Incubate at RT overnight

Check for clot lysis

Table 7. Two-tube technique interpretation for FXIII estimation

Thrombin+Urea Thrombin+Acetic acid Interpretation

+ + FXIII ≥ 18.8 IU/mL

+ - FXIII > 1 and < 18.8 IU/mL

- - FXIII < 18.8 IU/mL

Key: (-) clot lysed, (+) clot present

Mixing tests: These tests should be carried out with different proportions of patient and

normal plasma mixtures if clot solubility tests are positive to differentiate between a

congenital FXIII deficiency and acquired deficiency (presence of an inhibitor).

b. FXIII antigen estimation by ELISA

There are many ELISA kits with different working principle available in the market. In

routine laboratories generally the sandwich ELISA technique is employed wherein

monoclonal antibodies directed against each of the FXIII subunits i.e. FXIIIA and FXIIIB are

Page 329: Training Manual for Hemoglobinopathies and Hemophilia:

329

used as capturing and detecting antibodies. These tests detect only the FXIII tetramers; the

ones which are not complexed will be undetected. Plasma samples can be preserved in -700C

freezers and processed in batches. These ELISA assays are highly specific and sensitive.

One stage APTT-based factor assays (FVIII, FIX, FXI, and FXII)

For the measurement of factors VIII, IX, XI and XII, one stage APTT-based factor assay is

widely used. Factors X, V and II can also be assayed using this test, although for these

factors, a PT-based assay is more commonly used.

In hemophilia A patients, the severity of the bleeding disorder is depicted by the degree of

factor deficiency. Haemophilia patients are generally classified into three categories by their

factor activity: mild: 6-40% of normal; moderate: 1-5% of normal; and severe: < 1% of

normal.

Principle

The degree of correction obtained when the test plasma is added to specific factor deficient

plasma can determine the percent of factor present in the test plasma. APTT determines this

degree of correction. The results are compared to the degree of correction obtained when

normal plasma is added to same specific factor deficient plasma. Normal plasma is presumed

to give 100% correction.

Requirements

Citrated plasma of the patient

Standard plasma or normal pooled plasma (NPP)

Factor deficient plasma (substrate plasma): This is obtained either commercial or from a

hemophilic donor whose specific factor is <1 % of the standard plasma, absent for

inhibitors and transfusion transmitted diseases like HIV, HBsAg and HCV.

APTT reagent

0.025M CaCl2

Imidazole buffer pH 7.35 or Owren’s buffer/ Owren’s barbiturate saline (OBS), pH 7.35

i.e. barbiturate buffer pH 7.35 with 0.9% normal saline in 1:4 ratio

Procedure

Make serial dilutions (1:5, 1:10 and 1:20) of patient plasma and NPP in OBS or Imidazole

buffer

Diluted plasma (50 μL) + Factor deficient plasma (50 μL) + APTT reagent (50 μL)

Incubate for 3 minutes at 370C

0.025M CaCl2 (50 μL)

Page 330: Training Manual for Hemoglobinopathies and Hemophilia:

330

Normal range: 50- 150 % of NPP

The results are plotted on a semi-log graph paper with the time in seconds on the ordinate and

percent of dilution on the abscissa. By finding the point where the time obtained for the 1:5

dilution of the patient plasma intercepts the normal line, percent factor activity is read from

the abscissa of the graph. 100% factor is representation by the first dilution, i.e. 1:5 dilution

in this case, 1:10 represents 50% and 1:20 represents 25% factor of normal pooled plasma.

This assay is performed for factor VIII, IX, XI and XI assay using the respective deficient

plasma.

7. One stage PT- based factor assay (FVII)

For determining factor VII levels, a method based on PT is used. This same method can also

be employed for determining factor V, X and II levels.

Principle

The degree of correction obtained when the plasma is added to FVII deficient plasma helps

determine the percent of FVII present in test plasma. The PT determines the degree of

correction and this is compared to the degree of correction obtained when normal plasma is

added to the FVII deficient plasma. Normal plasma is presumed to give 100% correction.

Requirements

Citrated plasma of the patient

Standard plasma or NPP

FVII deficient plasma (substrate plasma): This is obtained either commercial or from a

FVII deficient donor whose FVII:C is <1 % of the standard plasma, absent for inhibitors

and transfusion transmitted diseases like HIV, HBsAg and HCV.

PT reagent.

0.025M CaCl2

Imidazole buffer pH 7.35 or OBS, pH 7.35

Procedure

Make serial dilutions (1:5, 1:10 and 1:20) of patient plasma and NPP in OBS or Imidazole

buffer

Diluted plasma (50 μL) + FVII deficient plasma (50 μL)

Incubate for 1 minute at 37°C

PT reagent (100 μL)

Normal range: 50- 150 % of NPP

Page 331: Training Manual for Hemoglobinopathies and Hemophilia:

331

100% factor VII is represented by the 1:5 dilution, 1:10 represents 50% and 1:20 represents

25% factor levels of normal pooled plasma. The graph is plotted in the same way as given

above in one stage APTT-based assay.

Either PT mode or APTT mode may be chosen using specific deficient plasma, for measuring

factors in the common pathway (factor II, V and X).

FVIII/FIX inhibitors

In patients having prolonged bleeding episodes or who do not respond well to the

conventional factor replacement therapy, inhibitors are generally suspected. Also before

surgical procedures, inhibitor screening tests are recommended so that the treatment products

can be kept handy. Patients once diagnosed with inhibitors need to be tested and monitored

periodically to ensure efficacy of treatment. Inhibitor screening assays are also advised when

patients are being treated with new factor products to check the immunogenicity of these

products. Immediate recognition of inhibitors is critical in most cases, since early therapy can

be life-saving.

When APTT is not corrected in mixing studies of a hemophilic patient plasma with NPP, it

indicates the presence of inhibitors. This could be against the coagulation factors or other

non-specific antibodies like LA. Patients could also have a heterogeneous mix of antibodies

which results in an accurate diagnosis.

Factor VIII / IX Inhibitor screening assay using NPP/FVIII concentrate

FVIII inhibitors are generally known to be time and temperature dependent i.e. detected after

incubation at 37°C over 2 hours. However, a heterogeneous mix of antibodies, including

lupus anticoagulants (LA) or even natural antibodies in NPP, could make diagnosis difficult

and give false-positive and non-specific results. Thus, using pure FVIII concentrates as the

‘control’, instead of NPP traditionally used to screen for FVIII inhibitors is more feasible. 1

IU/mL (100% FVIII: C) is prepared by diluting FVIII in Imidazole buffer pH 7.35. Instead of

NPP which is used to screen for FVIII inhibitors over 2 hours at 37°C, this FVIII is used. The

inhibitor screening protocol is as described below. The ‘incubated mix’ and ‘immediate mix’

APTT readings are noted and compared over 2 hours.

Reagents

Plasma-derived monoclonal purified FVIII or NPP

Test plasma

Reagents required for APTT test

Method

Page 332: Training Manual for Hemoglobinopathies and Hemophilia:

332

This screening assay involves incubating FVIII concentrate/ NPP, patient plasma and a 1:1

(or 3:1) NPP: patient plasma mix (incubated mix) in 3 tubes at 37°C for 2 hours. Most

importantly, to account for an increase in all the APTT readings after incubation at 37°C, the

APTT reading of the incubated mix is compared to that of a control immediate mix (1:1 mix

of the normal and patient plasma samples incubated separately). If the APTT reading of the

incubated mix is much higher than the immediate mix (generally >5 secs), the presence of an

inhibitor is indicated. This is subsequently confirmed by Bethesda assay.

Table 8: APTT- based Inhibitor Screen results of a positive sample

APTT 1 hour

APTT (seconds)

2 hour

APTT (seconds)

Incubated Mix 68 76

Separate Mix 58 60

Inhibitor screen test: Positive

Inhibitor Positive: Incubated Mix reading (seconds) > Immediate Mix reading (seconds).

However, this should be confirmed by Bethesda assay.

Inhibitor Negative: Incubated Mix reading (seconds) < Immediate Mix reading (seconds).

For screening for FIX inhibitors, same procedure is followed as for the Factor VIII

inhibitor assay by using NPP.

b. Classical Bethesda assay

This assay involves a two-hour incubation of a mixture of patient plasma with normal

pool plasma (NPP). The residual FVIII is compared with a similarly treated control

mixture containing buffer and NPP. This percent residual activity in the patient mix is

converted to Bethesda units (BU). One BU is defined as the amount of inhibitor which

will neutralize 50% of one unit of FVIII: C in normal plasma after 2 hours of incubation

at 37°C.

c. Nijmegen-modified Bethesda assay

The Nijmegen-modified Bethesda assay involves two modifications from the classical

Bethesda assay: i) To prevent loss of FVIII activity due to variation in pH, NPP is

buffered with 0.1 M imidazole pH 7.4 ii) factor deficient plasma is used in place of

Page 333: Training Manual for Hemoglobinopathies and Hemophilia:

333

imidazole buffer which helps to maintain constant protein concentration, thus

preventing any loss of FVIII activity.

Method

1. Doubling dilutions of patient plasma in FVIII deficient plasma is prepared. Depending

on the past or predicted inhibitor titer, the number of dilutions might vary.

2. Label standard tube and siliconized glass test tubes starting from 1:2,

1:4……….1:128….

3. Add 100 uL buffered NPP to all tubes. Add 100 uL FVIII deficient plasma to the buffer

blank tube and dilutions of patient plasma to the respective tubes.

4. Incubate for 2 hours at 370C

5. Place all tubes on ice at the end of 2 hours and measure residual FVIII activity in the

buffer blank standard and the first dilution. The first dilution residual FVIII assay will

provide a clue as which dilutions should be further analyzed.

6. That dilution of test plasma with residual FVIII closest to 50% of the buffer blank

standard is chosen for inhibitor titer calculation.

7. If the 1st dilution of test plasma and the second dilution of buffer blank standard do not

exactly match, the residual FVIII should be plotted in a log – log graph with FVIII in X-

axis vs inhibitor units in Y-axis. The actual FVIII inhibitor titer is calculated by

multiplying the residual FVIII with dilution factor.

Figure 5. The Bethesda assay graph (Dacie and Lewis 1999)

Page 334: Training Manual for Hemoglobinopathies and Hemophilia:

334

Buffering of NPP: Add solid Imidazole to NPP to make a final concentration of 0.1 M.

Adjust the pH to 7.4 slowly by adding 1N HCl with constant stirring at 40C.

ELISA assay for FVIII antibody detection

Below is the in-house method described by Shetty et al 2003 with minor modifications:

Reagents

Coating buffer consist of 15 mM phosphate, 150 mM NaCl, pH 7.4

Fixing solution consist of 0.05% Glutaraldehyde in coating buffer, immunoglobulin

standard.

Solid surface antigen consist of reconstituted lyophilized full-length recombinant FVIII in

coating buffer used at a concentration of 5U/well i.e. 5U/200µl.

Wash buffer contains phosphate 150 mM, 15 mM NaCl, 0.05% Tween 20, pH 7.4

Sample Dilution Buffer is 1% Gelatin in PBS.

Blocking solution is 3 % Gelatin in wash buffer.

Conjugate is human polyclonal IgG labeled with alkaline phosphatase.

Substrate solution consist of p-nitrophenyl phosphate (PNPP) 2 mg/ml in coating buffer.

Method

Pre- coat 96 wells flat bottomed ELISA plates with 0.05% glutaraldehyde overnight at 4°C

Wash with wash buffer 5 times

Coat the wells with 200 µL antigen i.e. recombinant FVIII

Cover tray, incubate overnight at 4°C

Wash with wash buffer 5 times

Remaining sites are blocked with 200 µL wash buffer with 3% gelatin for 1 hour, 20°C

Wash with wash buffer 3 times

Drain on absorbent paper

Add 200 µL of the test/ standard plasma in appropriate dilutions

Incubate for 2 hours (37°C)

Wash with wash buffer

Add 200 µL conjugated antibody which is diluted 1/10 times

Incubated for 2 hours

Wash with wash buffer

Incubate with 100 µL of substrate (PNPP)

Stop the reaction after 15 minutes using 3N NaOH

Page 335: Training Manual for Hemoglobinopathies and Hemophilia:

335

Take absorbance in an ELISA reader at 405 nm

Platelet aggregation tests

Principle

The adherence of one platelet to another is termed as Platelet aggregation. This phenomenon

can be induced by adding aggregating agents to PRP or whole blood where in the aggregation

depends on the presence of fibrinogen, Ca++, one or more plasmatic factors and an

aggregating agent. Each aggregating agent and their varying concentrations will give

different results in platelet aggregation test.

Page 336: Training Manual for Hemoglobinopathies and Hemophilia:

336

Procedure

Switch on aggregometer and recorder and wait for temperature display to indicate 37°C

Pipette 500 µL of test sample into the cuvette and add the stir bar

Pipette 500 µL of Reference (platelet poor plasma) into a second cuvette

Open the heater block cover and place the Sample into the well labeled PRP. Place the

Reference into well labeled PPP. Close the cover

After incubation for 2-3 minutes, add the agonist

Lower the recorder pens and switch “ON” the chart drive. Set the baselines by using the “Set

Baselines” button. Allow the chart to run for long enough to ensure the 0% (PRP) baseline

has stabilized

Open heater block and add appropriate agonist to the sample

Close heater block cover and allow the optical curve to run for the desired test time interval

At the end of the test time interval, raise the recorder pens and switch “OFF” the chart drive.

Platelet receptor studies

In this test platelets are labelled with fluorescent labeled monoclonal antibodies (Mab)

directed against surface membrane glycoproteins and then analyzed for their binding to

specific receptors by flow cytometry. It is a rapid and sensitive technique for diagnosis of

important platelet receptor defects such as Glanzmanns’s Thrombasthenia (GT) & Bernard

Soulier Syndrome (BSS). Whole blood or PRP can be used for testing. Additional tests for

confirmation may include gene sequencing to identify the causative mutation followed by

family studies.

Page 337: Training Manual for Hemoglobinopathies and Hemophilia:

337

Table 9. Fluorescent labelled antibodies and their specificities for the diagnosis of GT

and BSS

Mab Specificity

CD41 Platelet membrane glycoprotein GPIIb (the integrin alpha IIb chain) is recognized

by this antibody. GPIIb is non-covalently associated with GPIIIa (the integrin beta

3 chain) to form the GPIIb/IIIa complex.

CD61 The platelet membrane glycoprotein GPIIIa (the integrin beta 3 chain) is recognized

by CD61

CD42b GPIb on megakaryocytes and platelets are bound by CD42b. Ristocetin-dependent

binding of VWF to platelets and ristocetin-induced platelet agglutination is also

inhibited by CD42b

CD42a The platelet membrane glycoprotein GPIX is recognized by CD42a

Method:

Phosphate buffered saline (PBS) (50 μL) + PRP (5 μL) with platelet count about 250 x

109/L+ 2 μL of respective monoclonal antibodies

Tube 1: Unstained platelets

Tube 2: GPIX: Mouse anti-Human FITC/ PE tagged CD42a

Tube 3: GPIb: Mouse anti-Human FITC/ PE tagged CD42b

Tube 4: GPIIb: Mouse anti-Human FITC/ PE tagged CD41

Tube 5: GPIIIa: Mouse anti-Human FITC/ PE tagged CD61

Glanzmann's Thrombasthenia (GT): No positive shift of peak with either CD41 or CD61

(GPIIb/IIIa) due to the absence of GPIIb/IIIa complex is seen in GT patients, however a

normal peak with CD42b i.e. normal GPlb receptors is observed.

Bernard Soulier Syndrome (BSS): Normal binding with CD41 or CD61 is seen in BSS

patients, indicating a normal GPIIb-IIIa complex but no positive shift of peak with CD42 is

observed indicating the absence of the GPIb receptor.

Laboratory investigations for diagnosis of von Willebrand disease (VWD)

VWD is characterized by qualitative or quantitative deficiency of von Willebrand factor

(VWF). VWD can be classified into 3 main subtypes based on the type of defect;

Quantitative defects: type 1 and type 3, whereas type 2 is a qualitative defect. Type 2 VWD

can further be classified into 4 categories - 2A, 2B, 2M and 2N. The inheritance may be

autosomal dominant or recessive, depending on the subtype.

Page 338: Training Manual for Hemoglobinopathies and Hemophilia:

338

a. RIPA: Ristocetin induced platelet aggregation

The interaction between VWF and platelet glycoprotein Ib (GPIb) is promoted by Ristocetin,

which is an antibiotic. The procedure is explained under Platelet aggregation section.

A concentration of 1.25 mg/mL Ristocetin is used for the test. However if type 2B VWD is

suspected, a low dose ristocetin i.e. 0.5 mg/mL should be used for the test. Presence of

aggregation with low dose ristocetin is suggestive of type 2B VWD.

b. von Willebrand Factor Antigen (VWF:Ag) ELISA

A modified method followed by Dacie and Lewis 1999 is used. A microtitre plate is coated

with a primary antibody to VWF:Ag. A suitable dilution of the test plasma is added to these

wells; VWF:Ag binds to the primary antibody. By washing the plate, excess antigen is

removed; the second antibody conjugated to an enzyme, usually peroxide, is added which

binds to the VWF:Ag already bound to the plate. A color change occurs on addition of a

specific substrate. The reaction is stopped with an acid and the optical density (OD) of the

wells is measured using an ELISA plate reader. The OD is directly proportional to the

amount of VWF:Ag present in the test plasma.

Reagents

Microtitre plates

0.05 M Carbonate buffer: composition: 1.59g Na2CO3, 2.93g NaHCO3, 0.2g NaN3 in 1L

of D/W (pH 9.6)

Phosphate buffered saline: composition: 0.39g NaH2PO4.2H2O, 2.68g Na2HPO4.12H2O,

8.47g NaCl in 1L of D/W (pH 7.2)

0.01M Citrate phosphate buffer: composition: 8.8g citric acid, 24.0 Na2HPO4.12H2O in

1L of D/W

3% gelatin and 1% gelatin solution

Anti VWF:Ag antiserum (Diluted in 0.05M carbonate buffer)

Anti VWF:Ag conjugate with peroxide (Diluted in PBS-Tween-1% gelatin)

100% Calibration plasma

Test and control platelet poor plasma

1,2-o- Phenylenediamine dihydrochloride (OPD, Urea Peroxide)

Tween 20

1M HCL

Microtitre plate reader

Page 339: Training Manual for Hemoglobinopathies and Hemophilia:

339

Procedure

Add diluted anti-human VWF:Ag (100μL) to each well

Incubate for 1 hour in a moist chamber

Discard antibody; wash 3 times with PBS

containing 0.5 mL/L tween for 2 minutes

Invert onto an absorbent paper

Add 3% gelatin (100μL) as blocking agent

Incubate for 1 hour at 37°C

Prepare dilutions of the 100% standard plasma (1:10, 1:20, 1:40, 1:80, 1:160) in PBS with

1mL/L tween and 1% gelatin

Dilute patient and control plasmas (1:10, 1:20 and 1:40) in the same way

Add 100 μL of each dilution in duplicate to the wells

Incubate for 2 hours at 37°C

Repeat the washings

Add diluted anti-human VWF:Ag peroxide conjugate (100μL) to each well

Incubate for 2 hours at 37°C

Repeat washings as before and was once with 0.1M citrate phosphate buffer

Dissolve the OPD tablet in 20 mL D/W (freshly prepared) and add 100 μL to each well

Stop the reaction by addition of 50 μL of 1M HCL when the color has reached intensity

where a mid-yellow ring is clearly visible at the bottom of the wells

Read the OD at 492 nm

Plot a standard curve on log-linear graph paper and obtain the VWF:Ag levels by reading

from the reference curve

Normal range: 50-200 IU/ dL

c. Determination of VWF:Ag by LaurellImmunoelectrophoresis

Principle

Page 340: Training Manual for Hemoglobinopathies and Hemophilia:

340

Into an agarose plate, a precipitating rabbit anti-serum to human VWF: Ag is incorporated;

the wells of which are filled with test/ standard plasma. After applying an electric current;

precipitation peaks are obtained; the height of which is proportional to the concentration of

VWF:Ag in the samples. The test is insensitive for mild to moderate deficiency of VWF.

Requirements:

Agarose: Low electro-endosmosis

Barbitone buffer: Composition: 0.05 mol/L, pH 8.4 (17 g of Barbital-Natrium powder and

3.722 g of EDTA and add HCL to adjust pH 8.4 ; add D/W to make up the volume to 2

L)

NaCl 30g/L

EDTA- dipotassium salt

Polyclonal Rabbit Anti-Human VWF

Hydrochloric acid about 35% pure

Coomassie Blue stain

Coomassie Brilliant Blue (R): Composition: 5gms is added to Methanol 96% (450 mL)

and 100 mL of Acetic Acid glacial (100%). This is incubated overnight at room

temperature and add distilled water (450 mL). This solution is filtered before use.

Destainer: 250 mL of 96% methanol, 100 mL of acetic acid, 450 mL of distilled water

Calibration plasma (VWF - 100%)

Platelet poor plasma (tests and controls)

Apparatus: Glass Plate – 9 x 11cm, Water bath (56•C), Well borer (to give 2mm well size),

Whatmann filter paper no. 1, Wicks, Horizontal electrophoresis tank, Power pack

Method:

Dissolve 200 mg of agarose in 18 mL of 1:1 diluted Barbitone Buffer pH 8.4 by boiling in a

conical flask. Transfer to a 56°C water bath

Clean the glass plates with spirit to remove dust and grease

Add 18 µL of antiserum to the boiled agarose (cooled till 55°C) and mixed gently

Pour the agarose carefully and spread quickly on the plate to form an even film. Allow to

cool and after it is set, keep in a moist chamber at 4°C until ready for use

Cut equidistant wells in the agarose plate

Page 341: Training Manual for Hemoglobinopathies and Hemophilia:

341

Add 5.5 µL of the sample (fresh sample or thawed at 37°C) to each of the wells

Place the plates in a tank containing barbitone buffer. Place the wells on the cathode side so

that the samples migrate towards the anode. The electrophoresis is carried out at 16 mA for

18 hours

Remove the glass plates and immerse in 3% NaCl for 24 hours to elute out any excess

protein. Immerse in 0.9% NaCl and finally in D/W

Cover the plates with filter paper and keep in a hot air oven for drying

Coomassie Blue is used for staining for 5-10 minutes. Remove the excess stain by using

destainer. Allow the plates to dry.

Measure the height in mm from the middle of the well to the tip of the peak and compare

with the peak of the control sample or the calibrator and calculate the VWF values of the test

samples against the calibrator values.

Normal range: 50-150%

d. VWF:RCo: VWF Ristocetin cofactor assay

Principle

Unless normal plasma is added as a source of VWF, washed platelets do not ‘agglutinate’ in

the presence of Ristocetin. Agglutination follows a dose response curve which is dependent

upon the amount of plasma added. In this assay, freshly washed platelets or formaldehyde

fixed platelets can be used. Though it takes longer to prepare fixed platelets, these are not

susceptible to ristocetin aggregation, and they can also be stored for emergency use. It is

quicker to prepare freshly washed platelets and retain a functional platelet membrane, but

they cannot be retained for later use. Commercial lyophilized washed platelet preparations

are also available. Depending on the work load and urgency of the test, the type of platelet

preparation which best suits an individual laboratory can be decided.

Reagents

Sodium citrate solution. 32g/L trisodium citrate (Na3C6H5O7.2H2O)

K2EDTA. 0.134 mol/L

0.2% paraformaldehyde in 9g/L NaCl

0.05% Sodium Azide

Ristocetinsulfate: 1mg/mL reconstituted in distilled water.

Page 342: Training Manual for Hemoglobinopathies and Hemophilia:

342

Normal pooled plasma (NPP)/ standard plasma

Patients platelet poor plasma

Procedure

Obtain citrated blood from a normal subject with a normal platelet count

Centrifuge the blood at 300g for 15 minutes at RT

Separate the PRP and add 9 volume of PRP to 1 volume of EDTA solution, incubate for 1

hour at 37ºC to reverse the effect of ADP released during the preparation

Add an equal volume of 0.2% paraformaldehyde and leave at 4ºC. Decant the supernatant

and recentrifuge at 250g for 20 minutes at 4ºC

Discard the supernatant and resuspend the platelet sediment in chilled saline (9 g/L)

Wash the platelets twice

After the final wash, resuspend the platelets in sodium azide solution. Adjust the platelet

count to 300-500 Χ 109/L. The suspension is stable for 1 month at 4ºC.

Method

Add 495 μL of washed platelets & 1 μL of ristocetin in platelet aggregometer cuvette. Zero

agglutination of the platelets should be seen. The absorbance taken represents zero (%)

agglutination (blank); which should not exceed 5 divisions on the chart paper (5-6%

agglutination). If it is greater, then the platelets should be washed again and the above

procedure repeated. Repeat the reading of this blank after one hour

Add 500 μL of citrate saline into another cuvette and place in the warming block. This

reading represents 100% agglutination

Make serial dilutions from 1:2 to 1:32 of NPP in citrate saline

Similarly test the patient plasma in two dilutions, depending on the expected concentration of

VWF in the plasma. Both dilutions should give agglutination within the range of that of the

standard curve

Page 343: Training Manual for Hemoglobinopathies and Hemophilia:

343

To each cuvette add the following for the assay:

0.4 mL platelet suspension + 0.1mL plasma dilution + 5μL Ristocetin

Reset 100% and zero agglutination for each patient

Plot a graph of agglutination on the linear scale and the concentration of VWF on the log

scale on semi-log paper

Read the patient’s VWF level directly from the standard curve, correct for the dilution factor

and average of the two results from the different dilutions are taken.

Note: Addition of paraformaldehyde should be very slow with constant and gentle mixing of

the solution to avoid platelet clumping.

Normal range: 50-200 U/Dl

e. VWF:CBA- VWF: Collagen binding assay

Principle

The assay involves the direct binding of the plasma VWF to a solid phase antigen i.e.,

collagen, which is subsequently detected using anti-human VWF- HRP conjugate.

Reagents:

Microtitre plates

Phosphate buffered saline: 0.39g NaH2PO4.2H2O, 2.68g NaH2PO4.12H2O, 8.47g NaCl in

1L D/W (pH 7.2)

Collagen; pepsin digested type III collagen from human placenta 100 μg/mL i.e.

10μg/well is added.

Gelatin

Anti-VWF:Ag conjugate with peroxidase

100% calibration plasma

Platelet poor plasma (tests and controls)

1,2-o-Phenylenediamine dihydrochloride (OPD, Urea peroxide)

Tween 20

1M HCL

Microtitre plate reader

Washing Buffer: 0.1% Tween –PBS pH 7.2

Dilution Buffer: 1% Gelatin in PBS pH 7.2

Page 344: Training Manual for Hemoglobinopathies and Hemophilia:

344

Procedure:

Add 100 μL of collagen to microtitre plate

Incubate overnight at 4°C.

Wash the plates five times by washing buffer

Add 100 mL of 3 % gelatin as a blocking agent.

Incubate at 37°C; repeat the washings

Dilute the standard plasma i.e., NPP in 1:5, 1:10, 1:20, 1:40, 1:80. 1:160 and 1:320 and the

patient plasma into 1:5, 1:10, 1:20 and 1:40 dilutions in 1% gelatin

Add 100 μL of diluted plasma to each well

Incubate at 37°C for 2 hours followed by 5

washings with washing buffer

Add 100 μL of anti-human VWF-HRP conjugate

Incubated for 1 hour at 37°C

Add 100 uL of the substrate i.e,. 1,2-o-Phenylenediamine dihydrochloride (OPD) to each well

Incubation for 30 minutes

Add 50 μL of 1N HCL to stop the reaction

Measure the color developed in a microtitre plate reader at 492 nm

Plot the standardized curve on a log-linear graph paper. Three different curves are obtained.

1:5 dilution of the standard plasma is considered as 100% activity and activity is obtained by

reading from the reference curve.

f. VWF multimer analysis

Principle: von Willebrand factor circulates in the plasma as dimmers and multimers. For

subtyping the VWD, determination of these dimers and multimers is essential. The precise

sub-type of VWD is determined by electrophoretic separation and visualization of the

multimeric components of VWF binding with a radio-labeled anti-VWF antibody which is

observed by exposure to X-ray film.

Agarose gel electrophoresis

Reagents: Glycine, TRIZMA Base, Urea, Sodium dodecyl sulphate (electrophoresis grade),

Agarose (Ultra pure DNA grade), Bromophenol Blue (1% Stock Solution), Sodium EDTA.

Page 345: Training Manual for Hemoglobinopathies and Hemophilia:

345

2H2O, Deionized Water, Electrophoresis tank, Cooling System, 60°C water Bath, Glass

Plates, Spacers, Well cutters, Well template, Whatmann 3mm filter paper, Scalpel.

Procedure

Stock solutions:

2M TRIZMA base: 121.1g in 500 mL water. Stable at 4°C for up to 3 months

3M HCl

0.01M sodium EDTA. 2H2O: 1.86 g in 500 mL water. Stable at 4°C for up to 3 months

Running Buffer (x4 Conc.): 37.5mL 2M TRIZMA base + 4.0 mL 3M HCl. Make up to

50 mL with water. Adjust pH to 8.8 with 3M HCl. Store at 4°C for up to 2 weeks

Stacking Buffer (x 4 conc.): 12.5 mL 2M TRIZMA base + 7.0 mL 3M HCl. Make up to

50 mL with water. Adjust pH to 6.8 with 3M HCl. Store at 4°C for up to 2 weeks.

Sample Buffer: 500 mL 2M TRIZMA base + 10 mL 0.01M sodium EDTA. Make up to

100 mL with water. Store at 4°C for up to 2 weeks. [N.B. This is stock solution. Urea and

SDS are added to make the working solution.]

10% SDS: 1g SDS in 10 mL water. Store at 4°C for up to 3 weeks. Warm at 37

°C to

dissolve SDS before use.

Electrophoresis:

On the day of electrophoresis, the following reagents were prepared:

1. Electrophoretic buffer: Glycine: 57.6 g, TRIZMA Base: 12.0 g, SDS: 2.0 g. Dissolve

in 1.5L of water then make up to 2L. Before use, cool the buffer to 4°C.

2. Working sample buffer: Urea: 9.61 g, SDS: 0.40 g. Dissolve in the sample buffer by

warming at 37°C. Make up to 20 mL and allow to cool to room temperature. Adjust the pH

to 8.0 with 1M HCl, gently adding one drop at a time.

3. Mould the gel assembled by mounting a 1mm spacer on a clean glass plate. Mount the

second plate and line them up so that all the edges are aligned. Clamp all together with

bulldog clips, leaving the top open for pouring the agarose.

4. Preparation of gel:

Running gel: 1.6% agarose, 10% SDS

Agarose 0.4 g

Running buffer 6.25 mL

GDW 18.75 mL

Dissolve the agarose by heating for 30-35 seconds at 70% power levels in the microwave

oven. Stir this and reheat for 5-10 seconds to dissolve the entire agarose and add 250 mL of

Page 346: Training Manual for Hemoglobinopathies and Hemophilia:

346

10% SDS. Pour this carefully into the gap between the glass plates, allow to cool at RT for

about 15-20 minutes and then transfer to 4°C.

Stacking gel, 0.8% agarose, 10% SDS

Agarose 0.16 g

Stacking Buffer 5.0 mL

GDW 14.8 mL

Dissolve the agarose in the stacking buffer by heating to 100°C and once dissolved, add 200

μL of 10% SDS. Pour this into the gap between the glass plates above the running gel.

Allow to cool at RT for about 15-20 minutes, before transferring to 4°C. The excess stacking

gel is kept in the 60°C water bath for later use. When the gel has fully set, remove the clamps

and slowly remove the top glass plate by sliding off the gel. Wells are made in the stacking

gel with a well cutter.

Sample Preparation

Dilute the plasma samples as follows:

25 uL sample buffer + 25 uL sample + 7.5 uL 1% bromophenol blue

Incubate these in a 60°C water bath for 30 minutes and keep at 4

°C prior to electrophoresis.

Electrophoresis: Load 20 uL of diluted samples into the wells. Trasnfer the gel into the

electrophoresis tank with the wells on the cathode (negative electrode). Add approximately

600 mL of the electrophoresis buffer into each side of the tank. Place filter paper wicks on

both sides of the gel so that it is in even, parallel contact with the gel surface, but not in

contact with the wells. The electrophoresis is done under refrigeration at a constant current

of 8 mA. After about 1 hour, when the dye has left the wells, re-fill the molten stacking gel

into the wells. The samples are run for approximately 18 hours. When the electrophoresis is

complete, fix the gel and stain using the radio labeled probe.

Iodination of Antibodies: For radiolabelling the antibodies, Chloramine T method of

Greenwood et al 1963, modified by McConhey and Dixon 1966, is used. Take 1 mg protein

in 100 mL 0.1M PBS in a small round bottomed polysterene vial, in which 1mCi (3.7 x

107Bq) Na125I is added. Place a vial in a beaker containing ice to keep the reaction mixture at

low temperature. Prepare 10 mL of 1% Chloramine T in 0.1 M PBS (Azide free); add and

incubate for 3 minutes with constant shaking. Add another 10 mL and stop reaction at a total

of 10 minutes. Prepare 50 mL of 2% Sodium meta bisulfate in PBS and add. Run the

mixture through a Sephadex G-25 column, which is previously saturated with 200 mL of

20% bovine serum albumin (BSA). Elute thirty fractions of 0.5 mL each using PBS. Check

Page 347: Training Manual for Hemoglobinopathies and Hemophilia:

347

each fraction for serological titre and also radioactive counts per minute (cpm) of 10 mL

suspension in a Gamma counter. Two peaks are always obtained, the first peak having high

radioactive counts, contains the radiolabeled antibody and the second peak contains free

iodine absorbed by albumin, present in the column.

g. VWF:FVIII binding ELISA for the diagnosis of type 2 VWD Normandy (Type 2N

VWD)

The characteristic of Type 2 Normandy (2N) which is a qualitative defect of VWF, are low

FVIII:C levels (caused by a reduced affinity of VWF for FVIII), but normal VWD related

investigations. Mutations responsible for type 2N VWD are found in the FVIII binding

domain of VWF. Phenotypically, patients with type 2N VWD resemble patients with mild

hemophilia with reduced FVIII:C, and often have normal VWF:RCo and VWF:Ag levels.

Type 2N VWD has an autosomal recessive pattern of inheritance. The differential diagnosis

between hemophilia A and VWD type 2N has very important implications for both treatment

and genetic diagnosis.

VWF- FVIII binding assay is an ELISA-based method to determine whether VWF binds

normally to FVIII. VWF is captured using a monoclonal antibody; to remove endogenous

FVIII from VWF calcium chloride is used and a known amount of recombinant FVIII is

added to the bound VWF. Chromogenic FVIII assay is used to measure the bound FVIII.

This assay is similar to the one described below in the initial analytical steps. Microtitre wells

coated with rabbit anti-VWF antibody is utilized, which binds VWF/FVIII from diluted

patient plasma. After removal of endogenous (patient) FVIII, recombinant FVIII is added.

This binds to the patient VWF depending on the nature of the patient’s VWF molecule. The

only difference in the method from the one described below is the method of detection of

bound FVIII.

The general procedure of ELISA is as follows:

The VWF present in the test plasma is captured by the rabbit antihuman VWF F(ab’)2 which

is coated on the internal walls of a plastic microplate well. The FVIII in the tested plasma

dissociated during the first step is removed. The recombinant FVIII is then added which

binds to VWF. Next step involved binding of mouse monoclonal anti-human FVIII antibody

coupled with peroxidase to the remaining free antigenic determinants of the bound FVIII. The

bound enzyme peroxidase is revealed by its action on the TMB substrate. After stopping the

reaction with a strong acid, the intensity of the color is directly proportional to the

concentration of VWF bound to FVIII.

Page 348: Training Manual for Hemoglobinopathies and Hemophilia:

348

The ELISA process and the procedure may vary with different commercial sources. For a

good interpretation of VWF:FVIIIB results, a VWF:Ag level greater than or equal to 15% is

necessary. An over-estimation or an under-estimation of VWF:Ag level consequently leads to

an error of VWF:FVIIIB level.

Principle: The assay involves the direct binding of plasma VWF to a solid phase antigen i.e.

recombinant FVIII. This is subsequently detected by using anti-human VWF-HRP conjugate.

Reagents

Microtitre plates

0.05% Glutaraldehyde

Phosphate buffered saline: 0.39g NaH2PO4.2H2O, 2.68g NaH2PO4.12H2O, 8.47g NaCl in

1L D/W (pH 7.2)

Recombinant FVIII

Gelatin

Anti-VWF:Ag conjugate with peroxidase

Test and control platelet poor plasma

1,2-o-Phenylenediamine dihydrochloride (OPD, Urea peroxide)

Tween 20

1M HCL

Microtitre plate reader

Dilution Buffer: 1% Gelatin in PBS, pH 7.2

Washing Buffer: 0.1% PBS-Tween, pH 7.2

Blocking Buffer: 3% Gelatin in PBS, pH 7.2

Page 349: Training Manual for Hemoglobinopathies and Hemophilia:

349

Procedure

Add 100 μL of 0.05% Glutaraldehyde to microtitre plate

Incubate overnight at 4°C

Wash the plates 5 times with washing buffer

Add 100 uL of recombinant FVIII (12.76 U)

Incubate overnight at 4°C

Wash the plates 5 times with washing buffer

Add 100 mL of 3% gelatin as a blocking agent

Incubate at 37°C

Wash the plates 5 times with washing buffer

Dilute the standard plasma i.e., NPP into 1:5, 1:10, 1:20, 1:40, 1:80. 1:160 and 1:320

dilutions and the patient plasma into 1:5, 1:10, 1:20 and 1:40 dilutions in 1% gelatin

Add 100 μL of diluted plasma to each well

Incubate at 37°C for 90 minutes

Wash the plates 5 times with PBS- Tween

Add 100 μL of anti-human VWF-HRP conjugate (1: 2000)

Incubated for 1 hour at 37°C

Add 100 uL of the substrate i.e.OPD to each well

Incubate for 30 minutes

Add 50 μL of 1N HCL to stop the reaction

Measure the color developed in a microtitre plate reader at 492 nm

Plot the standard curve on a log-linear graph paper. Three different curves are obtained. By

reading from the reference curve, activity is obtained. 1:5 dilution of the standard plasma is

considered as 100 % activity.

(Adapted from Practical Immunohaematology edited by Shrimati Shetty; published by Jaypee Brothers)

Suggested Reading

Dacie JV and Lewis SM . Practical Haematology. Elsevier Publications 1999

Page 350: Training Manual for Hemoglobinopathies and Hemophilia:

350

Karpati L, Penke B, Katona E, Balogh I, Vámosi G, Muszbek L.A modified, optimized

kinetic photometric assay for the determination of blood coagulation factor XIII activity

in plasma.Clin Chem 2000;46:1946-55.

Kasper CK, Aledort L, Aronson D, Counts R, Edson JR, van EJ, Fratantoni J, Green D,

Hampton J, Hilgartner M, Levine P, Lazerson J, McMillan C, Penner J, Shapiro S,

Shulman NR. Proceedings: A more uniform measurement of factor VIII inhibitors.

ThrombDiathHaemorrh 1975;34:869–72

Kershaw G. Performance of Activated Partial Thromboplastin Time (APTT):

Determining Reagent Sensitivity to Factor Deficiencies, Heparin, and Lupus

Anticoagulants.Methods Mol Biol 2017;1646:75-83.

Mazurier C, Gaucher C, Jorieux S, Parquet-Gernez A. Mutations in the FVIII gene in

seven families with mild haemophilia A. Br J Haematol1997; 96:426–7.

Sadler JE, Ginsburg D.A database of polymorphisms in the von Willebrand factor gene

and pseudogene. For the Consortium on von Willebrand Factor Mutations and

Polymorphisms and the Subcommittee on von Willebrand Factor of the Scientific and

Standardization Committee of the International Society on Thrombosis and

Haemostasis.ThrombHaemost 1993 ;69:185-91

Shanbhag S, Shetty S, Kulkarni B, Ghosh K. An improved, semi quantitative clot based

assay for factor XIII.Haemophilia2011 ;17:718-20.

Shetty S, Ghosh K, Mohanty D.AnELISAassay for the detection of factor VIII antibodies

- comparison with the conventional Bethesda assay in a large cohort of haemophilia

samples.ActaHaematol 2003;109:18-22.

Shetty S, Ghosh K, Mohanty D.Comparison of four commercially available activated

partial thromboplastin time reagents using a semi-automated coagulometer. Blood

Coagul Fibrinolysis. 2003 ;14:493-7.

Verbruggen B, Novakova I, Wessels H, Boezeman J, van den Berg M, Mauser-

BunschotenE.The Nijmegen modification of the Bethesda assay for factor VIII:C

inhibitors: improved specificity and reliability.ThrombHaemost. 1995 ;73:247-51.

Yatuv R, Dayan I, Baru M. A modified chromogenic assay for the measurement of very

low levels of factor VIII activity (FVIII:C). Haemophilia 2006;12:253-7.

Page 351: Training Manual for Hemoglobinopathies and Hemophilia:

351

Genetic Counselling for hemophilia

Genetic Counselling is an integral part of a Comprehensive Hemophilia Care Center which is

also a major resource for patients and their relatives to discuss about all available options

within the existing resources. The whole process involves explaining the families of the

clinical severity, existing facilities for diagnosis and the various inheritance patterns, taking

maximum caution to adapt to the culture, religion and other circumstantial factor to which the

family is exposed to. Though for each disease , most of the information delivered to the

families are universally applicable across different countries, there are cultural, religious and

often individual barriers affecting the decisions of the families. Genetic counseling has an

important role even in the absence of genetic testing.

Components of genetic counselling process

The genetic counselling process involves the following important components

Developing a rapport with the family: This is the first and the most important step of a

genetic counseling session. Patients or family members express a wide range of feelings

which include fear, anxiety and guilt. It is the responsibility of the Genetic Counselor to

convince and counsel in such a way that they feel free to express their feelings.

Education :Prior to the counseling session, it is important to understand whether the family

has an understanding about the disease, management , course of the disease, existing

treatment products and so on, since this may have an impact the decision of the family.

Patients and families should be provided with complete information which will help them

through the genetic counseling procedure and the decision-making process subsequent to a

new diagnosis of hemophilia or diagnosis of a carrier

Pretesting Education

Identify person to be tested

Do they want to be tested and know results?

What does being a carrier mean ?

How is haemophilia inherited ?

What does the test involve ?

Family planning

Possible options for pregnancies in case of a carrier

Page 352: Training Manual for Hemoglobinopathies and Hemophilia:

352

Figure 6. Inheritance patterns in haemophilia

Page 353: Training Manual for Hemoglobinopathies and Hemophilia:

353

Once the family decides to go through the genetic diagnostic process, the first step is to draw

an extensive family pedigree to know the relationship of the potential carriers in the family

with the index case. To get a clear picture, this should always be drawn in the form of a

pedigree chart rather than in descriptive terms.The obligate carriers will be identified by

drawing the family pedigree itself. All the remaining potential carriers may be assigned

carrier or noncarrier status by subjecting them to genetic diagnosis if the family decides to do

so.

Carriers of hemophilia face quite a few important and tough choices. They can accept the risk

of having a child with haemophilia, or perform prenatal diagnosis with the associated

problem of termination of pregnancy in case the fetus is found to be affected. A few carriers

do not go for prenatal diagnosis because they do not see haemophilia as a life threatening

disorder to justify termination. Religion, culture and the quality of life of haemophilia

patients in the family may be the key decisive factors . A diagnosis of hemophilia or carrier

may certainly confuse the patient or the relatives. At this stage the Counselor should provide

various alternate options available such as preimplantation diagnosis, noninvasive procedures

and the final decision should be taken by the family

Informed consent : Informed consent for genetic testing from the patient should be obtained

before any procedure if the family is interested in going ahead with carrier diagnosis and/or

prenatal diagnosis. It is always better to assign this responsibility to the person who has done

the genetic counseling

Post-procedure counseling

Results

Carrier or not

Children or not

Natural conception or IVF/PGD

When and options to test the pregnancy

Keep pregnancy or terminate

Frequently asked questions

1. Who are called “ Obligate Carriers”?

Ans : The obligate carriers are i. Daughters of hemophilic patients ii. A lady with two

children with hemophilia iii. A lady having onechild with hemophilia and another

patient in the maternal side. Such carriers do not need any genetic investigations and

the family pedigree itself will assign them the carrier status.

Page 354: Training Manual for Hemoglobinopathies and Hemophilia:

354

2. If a lady has an affected brother and two normal sons , is there a chance of her being a

carrier ?

Ans : Yes, Since a carrier has one normal X chromosome and one affected X

chromosome, even if she is a carrier, both her children might have inherited only

normal X chromosomes. However, this does not rule out the possibility of her being a

carrier.

Can there be a child with haemophilia in the family without any family history?

Ans : Yes, around 40% of the families do not have a family history. There are three

possibilities ; either the mutation might have arisen in the germ cells of the maternal

grand father , which is transmitted to the mother and then subsequently to the son or the

family history may still be positive, but due to female preponderance in the family or

due to chance factors , the hemophilic phenotype might not have been shown up in the

family. The last possibility is of a true “ de novo” mutation, where the mother may not

be a carrier and the mutation might have occurred in the affected child only. Under

these circumstances any indirect method of genetic diagnosis becomes highly

inaccurate.

3. During antenatal diagnosis , can the carrier status of the fetus be informed?

Ans : No. The carrier status of the fetus is not detected antenatally in a family with

hemophilia. The laboratory reports indicate the type of mutation in the index case and

just mentions whether the fetus is “ affected” or “ unaffected”. The “affected”fetus

means that the child has hemophilia. The “unaffected fetus” means normal male,

normal female or carrier female.The carrier status in case of a female child will only be

determined after birth.

4. Are we able to provide an accurate diagnosis in all the 100% of the families referred for

genetic diagnosis?

Ans : With direct identification of mutations, the reports are highly accurate; however

there are certain inherent problems in haemophilia genetics like somatic and germ line

mosaicism, wherein one can miss the mutation even by direct DNA sequencing.

However this is rare. In 1-2% of the families, the mutations are not found in the coding

regions, which are the regions generally scanned by the conventional DNA sequencing

techniques. The reasons may be the presence of the mutation in the deep intronic region

. Only the advanced next generation sequencing techniques can detect such variants

which cause the hemophilia phenotype.

Page 355: Training Manual for Hemoglobinopathies and Hemophilia:

355

5. Can the factor VIII or IX levels determine the carrier status of a female ?

Ans : No, there is a wide range of factor levels both for carriers and noncarriers and

there is a substantial overlap in the factor levels between these two groups. So factor

levels cannot be used to assign “ carrier” or “ noncarrier “ status to a lady belonging to

a hemophilia family

6. If the index case is not available, can a carrier diagnosis or prenatal diagnosis be done

in the family?

Yes, With the availability of direct mutation identification techniques, both carrier

diagnosis and prenatal diagnosis can be done even in the absence of affected member.

7. At what age should the carrier testing be done in a female belonging to a hemophilia

family?

Ans : There is no age limit as when a female should be tested. However, the families

are informed about the availability of genetic diagnosis techniques and counseled to get

the carrier status confirmed before getting married or before conception, in case the

family is opting for subsequent antenatal diagnosis .

Suggested Reading

Alabek M, Mohan R and Raia MH. Genetic Counselling for Hemophilia . WFH

special edition

Miller R . Genetic Counselling for Hemophilia . WFH special edition 2002.No 25

Page 356: Training Manual for Hemoglobinopathies and Hemophilia:

356

Genetic Diagnosis of Hemophilia

Hemophilia A: Factor VIII deficiency

Factor VIII gene (F8) mutations are highly heterogeneous and are not restricted to any

specific region. F8 is one of the largest genes in human genome with 26 exons and 25

introns, with the coding region being 9 kb in size. The exons range in size from 69 bp (exon

5) to 3106 bp ( exon 14), whereas the introns vary in size fr

om 200 bp (intron 17) to 32.4 kb ( intron 22).

Figure 7. General strategy for genetic diagnosis in severe hemophilia A

Linkage analysis or restriction fragment length polymorphism (RFLP) analysis can also be

applied for genetic diagnosis. However it is no longer recommended.

Inverse-PCR Technique for Intron 22 Inversion Analysis

The method is described by Rossetti et al 2005 in detail and involves:

Day 1: Stage 1: Digestion of genomic DNA with Bcl1

Page 357: Training Manual for Hemoglobinopathies and Hemophilia:

357

i. 0.5-2 µg of extracted genomic DNA is added to a labeled 1.5 mL microvial tube in a

total volume of 15 µL with sterile D/W.

ii. A Bcl1 restriction digestion master mix is prepared for the required number of

samples according to the calculation for 1X reaction:

iii.

Reagents 1x Volume

(uL)

D/W 5.0

Bcl1 10X buffer 2.5

Bcl1enzyme 2.5

Total 10

iv. 10 µL of the above Bcl1 master mix is added to each sample.

v. Vortex to mix well, spin briefly in a microfuge.

vi. Incubate at 55oC in a water bath for a minimum of 5 hours (maximum incubation

period overnight).

Stage 2: Ligation

i. Digestion reaction from water bath is removed and briefly spun in a microfuge.

ii. T4 DNA ligase master mix is prepared for the required number of samples according

to the calculation for a 1X reaction:

Reagents Volume 1x (uL)

D/W 199

10X T4 DNA ligase buffer 25

T4 DNA ligase(5U /mL) 1

Total 225

iii. Vortex to mix well, spin briefly in a microfuge.

iv. Incubate at 15oC in a cool block (thermal cycler) overnight.

Stage 3: Concentration or precipitation of the B-rings:

Following the ligation reaction the products have to be concentrated:

i. The reaction tubes are briefly spun in a microfuge.

ii. The entire volume is transferred to a labelled Ultracel YM-100 spin column sat in a

1.5mL tube (as supplied with the columns).

iii. Spin at 8,000rpm for 8 minutes.

iv. The spin column are inverted into a new, labelled tube (as supplied with the columns).

Page 358: Training Manual for Hemoglobinopathies and Hemophilia:

358

v. Spin at 12,000 rpm for 4 minutes to recover the product from the column filter.

or

i. 30 µL of 3M sodium acetate and 800 µL of chilled ethanol is added to the entire volume

of each ligated sample in a 1.5 mL microvial tube and gently mix by inverting the tubes.

ii. Incubate the tubes at -20°C for 1 hour.

iii. Centrifuge the tubes at 14000 rpm for 20 mins to recover the DNA rings.

iv. The pellet obtained was air dried and 15 µL of D/W was added and allowed to dissolve at

room temperature overnight.

Stage 4: PCR amplification:

This stage must be performed in the PCR laboratory. The complementary PCR is an optional

test, which is not required as a first line test for the investigation of males with severe

haemophilia A.

i. In a 1.5 mL tube a PCR master mix is prepared containing all components of the PCR

reaction except the ligated DNA. Volumes stated below are for 1 reaction. Sufficient

should be made for all reactions + 1 blank (negative control) +1 to allow for pipetting

variance.

ii. The PCR amplifications are performed in a reaction volume of 25 µL with 11 µL sterile

D/W, 2.5 µL of 10x complete buffer, 0.3 µL of 25mM dNTPs, 1.25 µL of each Primer

(10pM) and 0.2 µL of DNA Taq polymerase (5U/µL).

iii. For the Diagnostic PCR, Primers 1U, 2U, 3U and ID are used and for the Complementary

PCR, 1U, 2U, 3U and ED primers are used in the concentrations mentioned above.

iv. Vortex to mix and transfer 19 µL to each labeled 0.2 µL PCR tube

v. Add 6 µL of diluted ligation product, or 6 µL deionised water to the negative control

tube.

vi. Transfer to a PCR machine in the main laboratory and run the following conditions:

94◦C- 12 minutes X 1 cycle

94◦C- 30 seconds, 57◦C- 1 minute, 72◦C- 1 minute X 30 cycles

72◦C- 5 minutes X 1 cycle

4◦C- ∞

Approx. time taken for amplification: ~2.5 hours

Stage 5: Post PCR:

i. Post-PCR samples can be stored at 4oC prior to analysis if required. Samples are analyzed

by transferring 15 µL of PCR product into a clean 0.5 mL tube. 3 µL of loading buffer is

then added.

Page 359: Training Manual for Hemoglobinopathies and Hemophilia:

359

ii. Samples are loaded onto a 2% agarose gel in 1x TAE buffer (100ml agarose gel in), with

a 100 bp DNA size marker. The gel should be prepared with a single comb at one end to

provide the maximum run length.

iii. Gels are run at 80V for to allow clear separation of PCR products till the bands separate.

iv. Results are visualized using the gel imaging system, and a photograph is taken.

Figure 8. Intron 22 inversion analysis by complementary test

(Adapted from Practical Immunohaematology edited by Shrimati Shetty; published by Jaypee Brothers)

Figure 9. Intron 22 inversion analysis by Diagnostic test

(Adapted from PracticalImmunohaematology edited byShrimati Shetty; published by Jaypee Brothers)

Page 360: Training Manual for Hemoglobinopathies and Hemophilia:

360

Table 10. Expected Results in the diagnostic and complementary tests

Alleles

Diagnostic test

ID + IU + 3U +

2U

(bp)

Complemetary

test

ED + IU + 3U +

2U

(bp)

Male

Wildtype (N) 487 457 + 405

Int-22 inversion type1 (Inv22-1) 333 559 + 457

Int-22 inversion type 2 (Inv22-2) 385 559 + 405

Benign duplication F8 1-22 (Dup1-

22)

487 559 + 457 + 405

Int-22 deletion type1 like (del22-1) 333 457

Int-22 deletion type2 like (del22-2) 385 405

Female

N/N 487 457 + 405

Inv22-1 /N 487 + 333 559 + 457 + 405

Inv22-2 /N 487 + 385 559 + 457 + 405

Dup1-22 / N 487 559 + 457 + 405

Del22-1 / N 487 + 333 457 + 405

Del22-2 / N 487 + 385 457 + 405

Poor quality DNA may give non-specific banding, which could be misinterpreted as

heterozygosity. Non-specific amplification can be clearly differentiated from true

heterozygosity with experience. Unusual banding patterns have been observed, which may be

benign or pathogenic in nature. Aberrant results should be further explored by additional

analysis of the F8 gene by MLPA and /or sequencing.

Intron 1 inversion Analysis

Intron 1 inversion is analyzed by the method described by Bagnall et al 2002.

Page 361: Training Manual for Hemoglobinopathies and Hemophilia:

361

PCR amplification of F8int1 regions and int1h repeats: 100 ng genomic DNA in a reaction

volume of 25µL with 2.5 μL 10 X buffer with MgCl2 (complete buffer), 1.5 mM MgCl2, 2

µM of each oligonucleotide primer, 25 mM of each dNTP, and 0.7 U DNA Taq polymerase.

Six primers are used in two sets for the multiplex PCR; group A: Primers 9F, 9cr and Int1h-

2F and group B: Primers Int1h-2F, Int1h-2R and 9F

Thermal cycling conditions

94°C- 5 minutes X 1

94°C- 30 seconds, 65°C- 40 seconds, 72°C- 2 minutes X 30 cycles

72°C- 5 minutes X 1

4°C- ∞

The PCR amplicons are analyzed on a 2% agarose gel.

Interpretation: The sample is normal in case, a 1.5 kb band is obtained in int1h-1 and a 1kb

band is obtained in int1h-2. The sample has intron 1 inversion mutation in case a 1 kb band is

obtained in int1h-1 and a 1.5 kb band is obtained in int1h-2. In a carrier both these bands are

observed in both the PCR tubes.

Figure 10.Intron 1 Inversion Analysis

(Adapted from Practical Immunohaematology edited by Shrimati Shetty; published by Jaypee Brothers)

Analysis of F8 mutations

The 26 exons of F8 may be scanned by 37 PCR reactions as given below:

Page 362: Training Manual for Hemoglobinopathies and Hemophilia:

362

Page 363: Training Manual for Hemoglobinopathies and Hemophilia:

363

Table 11. PCR reagent composition and PCR conditions for various exons of F8

Exon D/W

uL)

10x Incomplete buffer

(uL

Forward and reverse primers

(uL)

25 mM

MgCl2

(uL)

25 mM

DNTPs

(uL)

Taq DNA polymerase 5U/uL

(uL)

Annealing temperature

(°C)

Size (bp)

Pro1 29.6 5 3 8 0.6 0.4 54 540

Pro2 30.1 5 3 7.5 0.6 0.4 58 626

1 20.6 5 2 7 0.6 0.4 53 524

2 19.4 5 2.5 6.2 0.6 0.4 55 331

3 20.6 5 2 9 0.6 0.4 52 346

4 19.4 5 4 8 0.6 0.4 53 319

5 20.6 5 2 7 0.6 0.4 53 274

6 26.1 5 2 5.5 0.6 0.4 55 423

7 20.6 5 2 7 0.6 0.4 53 450

8 24.6 5 2.5 10 0.6 0.4 53 547

9 19.4 5 2.5 6.2 0.6 0.4 55 416

10 29.6 5 2 6 0.6 0.4 54 347

11 20.6 5 3 9 0.6 0.4 52 445

12 25.4 5 1.5 6.2 0.6 0.4 58 320

13 29.6 5 2 9.4 0.6 0.4 49 474

14A 27.6 5 2 7.5 0.6 0.4 55 518

14B 25.4 5 1.5 6.2 0.6 0.4 58 499

14C 19.4 5 2 6.2 0.6 0.4 55 499

14D 27.6 5 2 8 0.6 0.4 54 392

14E 25.4 5 1.5 6.2 0.6 0.4 58 501

14F 22.1 5 2 7.5 0.6 0.4 55 381

14G 27.6 5 2 8.5 0.6 0.4 54 440

14H 27.6 5 2 7.5 0.6 0.4 55 345

14J 29.6 5 2 6 0.6 0.4 54 483

Page 364: Training Manual for Hemoglobinopathies and Hemophilia:

364

14K 27.6 5 2 8.5 0.6 0.4 54 347

15 19.4 5 2 6.2 0.6 0.4 55 348

16 26.1 5 2 5.5 0.6 0.4 55 525

17 19.4 5 4 8 0.6 0.4 53 491

18 22.1 5 2.5 7.5 0.6 0.4 55 412

19 22.1 5 2.5 7.5 0.6 0.4 55 341

20 27.6 5 2 8 0.6 0.4 54 311

21 29.6 5 2.5 8 0.6 0.4 58 168

22 20.6 5 2 9 0.6 0.4 52 280

23 26.1 5 2 5.5 0.6 0.4 55 349

24 20.6 5 2 7 0.6 0.4 53 350

25 24.6 5 2 10 0.6 0.4 53 373

26 25.4 5 1.5 6.2 0.6 0.4 58 356

Page 365: Training Manual for Hemoglobinopathies and Hemophilia:

365

Table 12. Thermal cycling conditions for PCR amplification of various exons of F8

Denaturation Denaturation Annealing Extension Extension Final

Hold

F8 94°C

5 minutes

30 cycles

72°C

5 minutes

4°C

∞ 94°C

40 seconds

Annealing

temperature °C

( given in Table

below)

40 sec

72°C

40 seconds

Haemophilia B

Factor IX gene (F9) is approximately 33 kb in size with 8 exons and 7 introns with mRNA

being 2803 bp in size. The exons range in size from 25 bp (exon 3) to 1935 bp (exon 8). The

introns vary in size between 188 bp (intron 2) and 9473 bp ( intron 6)

For application of any screening test for identification of mutations, two or more exons with

more or less similar annealing temperatures and other parameters may be multiplexed to

make the technique cost effective.

Reagent Mix (50 μL) for a single PCR reaction for all exons of F9.

Exons 1 2+3 4 5 6 7 8A 8B PolyA

Annealing Temperature (°C) 54 57 62 57 62 54 50 57 54

10X PCR buffer (µL) 10 10 10 10 10 10 5 10 10

Forward Primer (µL) 2.5 2.5 2 1 2 1.5 1.5 2 1.5

Reverse Primer (µL) 2.5 2.5 2 1 2 1.5 1.5 2 1.5

25mM MgCl2 (µL) 2 1.5 1 1.5 1 2 2 1.5 2

25 mM dNTP (µL) 1

Taq Polymerase (µL) 1.5

D/W (µL) 29 29 30.5 31 30.5 31 37 30 30.5

DNA (µL) 2

Thermal Cycling conditions for all exons

94◦C- 5 minutes X 1 cycle

94°C- 40 seconds, annealing temp, refer table -40 seconds, 72°C- 40 seconds X 30 cycles

72°C- 5 minutes X 1 cycle

4°C- ∞

Page 366: Training Manual for Hemoglobinopathies and Hemophilia:

366

Women with bleeding disorders: Approach and laboratory diagnosis

It is important to recognise and diagnose bleeding disorders in women. The management of

bleeding would be most accurate when the etiology of the bleed is identified so that: Factor

replacement in factor deficiencies, platelet infusions for platelet factor defects and

antifibrinolytics in certain situations can be prescribed.

Important issues related to bleeding disorders in women-

a) Management of heavy menstrual blood flow especially in our country where adequate

provision of sanitary napkins is not a norm, leading to increased school dropout rates.

b) Worsening of already existing Iron deficiency anemia in women of poor socioeconomic

status and its attendant effects, which forms a bulk of the problem.

c) Life threatening bleeds in pregnancy and delivery with poor outcomes of unsupervised

pregnancies.

d) Increased load on a “stretched out” blood bank services.

e) Coexistent comorbidities may mask the bleeding to be a sign of a bleeding disorder and

delay its diagnosis.

Bleeding disorders that affect women

1) Von Willebrand Disease – Commonest coagulation disorder with varied phenotype –

presenting as mucosal wet/ dry bleeds

2) Haemophilia A and B (carriers generally do not bleed; however , few carriers have low

factor levels and they need to be investigated).

3) Platelet function disorders

4) Rare factor deficiency– Factor X, II, V, VII, X, XI, XIII

5) Collagen vascular diseases

Approach to a female with bleeding

A) Detailed history of bleeding

B) Family history

C) Laboratory tests

History of bleeding

Joint bleeds are infrequent and seen sometimes in severe VWD.

Bleeding in early life may indicate an inherited bleeding defect.

In infants – Intracranial bleeds, umbilical stump bleeding

In girls - epistaxis, easy bruising and dental bleeds

Page 367: Training Manual for Hemoglobinopathies and Hemophilia:

367

In adolescents and women – Menorrhagia, pregnancy related bleeds besides the other

bleeding complications

At any age- Post traumatic and post-surgical bleeds

Severity assessment can be made by using Bleeding Assessment Tools (BAT) or

questionnaires. Unlike the haemophilias in males, severe deficiency of other coagulation

factors (Factor II, V, VII, X XIII and fibrinogen) may not result in spontaneous bleeds,

thereby further delaying their diagnosis.

Comorbidities like liver disease, DIC, Vitamin K deficiency may confound the clinical

picture and results of tests, hence screening for these in the appropriate clinical setting is

advisable prior to undertaking extensive testing.

History of recent blood product therapy should be elicited since they can confound

laboratory results and mask an underlying severe defect.

Family history of bleeding

Most of the bleeding disorders in females are of autosomal recessive inheritance, wherein the

parents may be asymptomatic. Type 2 B vWD is inherited in an autosomal dominant fashion.

Carriers of X linked disorders such as Hemophilia A, may at times become symptomatic and

experience heavy bleeding and have low Factor VIII levels. This is the result of extreme

lyonisation of the normal X chromosome.

Family history may not be informative since current norms of bearing few children, results in

affected carriers not having any affected offspring by a turn of chance.

Laboratory Tests

Since most laboratory test for bleeding disorders are expensive a step wise screening and

confirmation is advisable.

1) Screening tests

2) Confirmatory tests

3) Specialised tests

4) Genetic tests

Page 368: Training Manual for Hemoglobinopathies and Hemophilia:

368

Screening tests

These tests will indicate if there is a coagulation or a platelet defect. These tests are sensitive

but not specific. The usual panel comprises

i) Prothrombin time (PT) – prolonged in Factor II, V, VII, X and fibrinogen deficiency

(in isolation, combination or as a part of liver disease, Vitamin K deficiency)

ii) Activated Partial Thromboplastin time (APTT)- prolonged in Factor II, V, X, VIII,

IX, XI, VWD deficiency

iii) Thrombin Time (TT) - Prolonged in fibrinogen deficiency or abnormality

iv) Fibrinogen assay- decreased in afibrinogenemia and hypofibrinogenemia

v) Platelet Count- decreased in Immune thrombocytopenias, DIC, Type 2BVWD,

Platelet Type vWD and some inherited macrothrombocytopenic syndromes

vi) Bleeding time – Prolonged in quantitative and qualitative platelet defects, VWD

vii) Urea clot solubility assay –prolonged in Factor XIII deficiency

viii) Platelet aggregation studies by light transmission aggregometry (LTA) – for

qualitative platelet defects – Glanzmann’s thrombasthenia , Bernard SoulierSyndome

viii) Screening for VWD- VWF antigen assay, VWF GPIbR assay, VWF Ristocetin

Cofactor assay, VWF Collagen Binding assay

Prolongation of the PT and APTT are followed by mixing studies with equal volumes of

normal plasma to establish a factor deficiency and exclude inhibitors.

The mixing tests can be modified to establishing the deficient factor by demonstrating failure

of normalisation of the clotting time on addition of individual deficient plasmas

Confirmatory tests

These tests are complex and expensive and are performed at tertiary centres with trained

manpower.

1) Once the deficient factor is identified then the degree of deficiency can be quantitated by a

specific factor assay e.g.- One stage clot-based Factor V assay for Factor V deficiency

2) Factor XIII assay by ammonia release methods

Specialised tests

These tests need expensive equipment and trained operators.

1) Platelet flowcytometry for CD41, CD6 and CD 42b, CD36 for confirming Glanzmann’s

thrombasthenia, Bernard Soulier syndrome, Collagen receptor defect

2) Electron microscopy – For confirming a decrease in the dense or alpha granule content in

storage pool disorders of platelets

Page 369: Training Manual for Hemoglobinopathies and Hemophilia:

369

3) vWF multimer analysis for confirming the subcategory of VWD

4) vWF / Factor VIII binding assay- For vWD Type 2N

5) Ristocetin induced platelet aggregation ( RIPA ) – For vWD Type 2B and Platelet Type 2B

6) VWF propeptide assay- for excluding acquired vWD in older females with bleeding

symptoms

Molecular tests

In women with bleeding disorders, molecular tests may be required to

1) Confirm the defect in cases where the phenotype of two disorders may be indistinguishable

on the currently available tests. The management of the two may be significantly different

e.g.- Type 2B VWD and Platelet Type VWD

2) For genetic counselling discuss the inheritance and likelihood of disease in offspring and

other related family members

3) For carrier screening and antenatal diagnosis in disorders associated with significant

bleeding

4) To understand the genetic basis of phenotypic variation in patients with similar diseases

Sequencing of the affected gene may be required for establishing the diagnosis. DNA

analysis of the family members is required to confirm the pattern of inheritance and provide

genetic counselling.

The acquired causes of bleeding especially liver disease, renal disease, DIC, hormonal

imbalance are far more common than any of the inherited bleeding disorders. While

investigating bleeding in women other than the tests specific for bleeding disorders it is

important to also correlate with results for the following tests to exclude the presence of a

comorbidity which may aggravate the bleeding.

1) Complete Blood count- For haematological causes for bleeding- aplastic anemia,

leukemia, DIC and for evidence of Iron deficiency

2) Examination of the peripheral blood film – Schistocytes in DIC, blasts in leukemias, giant

platelets in Bernard Soulier syndrome, washed out platelets in Grey Platelet Syndrome,

Nuclear inclusions in May Hegglin anomaly

3) Liver function tests

4) Renal Function test

5) Hormonal assays - Thyroid profile

6) Imaging studies- To exclude anatomical lesions e.g.- fibroids, polyps for menstrual

bleeding.

7) Tests for autoimmune disorders – Ds DNA

Page 370: Training Manual for Hemoglobinopathies and Hemophilia:

370

Hematochezia, hemoptysis and hematemesis are usually never associated with inherited

bleeding defects. Testing during pregnancy and puerperium may yield fallacious results since

levels of Factor VIII and VWF are raised in this time. Repeat testing is mandatory to avoid

misdiagnosis.Levels of Factor VII, V, IX, X are low in early infancy and may achieve

normal values only after 6 months of age. Hence the results of the girl child investigated in

infancy for these disorders must be compared with age related norms and reconfirmed again

after 6 months.Some polymorphisms in the VWF gene are associated with defective

Ristocetin binding but no bleeding symptoms. Some women may have low vWF antigen

levels with no bleeding.Hence the presence of an abnormal test should not be enough to label

a woman with a bleeding disorder in the absence of history of bleeding.

Page 371: Training Manual for Hemoglobinopathies and Hemophilia:

371

Table 13: Brief summary of tests in women with bleeding disorders

Disorder PT APTT Fibrinogen

assay

Platelet

Count

Thrombin

time

Mixing

studies with

normal

plasma

Factor assay

for

confirmation

Platelet

aggregation

Platelet flow

cytometry

Additional

assays Genetic testing

Von Willebrand

disease N N/↑ N N/↓ N ND

Factor VIII- N/↓

VWF antigen-

N /↓

VWF Ristocetin

cofactor

activity-↓

↓ with

Ristocetin N

VWF FVIII

binding,

VWF CBA,

RIPA,

Multimer

analysis,

Propeptide

assay

Y

Factor VIII/IX

carrier N ↑ N N N Correction FVIII/FIX ND ND ND Y

Factor II

deficiency

↑ ↑ N N N Correction Factor II assay ND ND ND

Genetic testing may be

required if married within

family

Factor V

deficiency

↑ ↑ N N N Correction Factor V assay ND ND ND

Genetic testing may be

required if married within

family

Factor X

deficiency

↑ ↑ N N N Correction Factor X assay ND ND ND

Genetic testing may be

required if married within

family

Factor VII

deficiency ↑ N N N N Correction Factor VII assay ND ND ND

Genetic testing may be

required if married within

family

Page 372: Training Manual for Hemoglobinopathies and Hemophilia:

372

Factor XI

deficiency

N ↑ N N N Correction FXI assay ND ND ND

Genetic testing may be

required if married within

family

Factor XIII

deficiency N N N N N NR FXIII assay ND ND

FXIIIA,

FXIIIA2

B2 assay

Genetic testing may be

required if married within

family

Afibrinogenemia,

hypodysfibrinogen

emia

↑ ↑ ↓ N ↑ ND Fibrinogen

assay NR NR

Antigenic

assay for

hypodysfibr

inogenemia

Genetic testing may be

required if married within

family

Glanzmann’s

thrombasthenia N N N N N ND ND

↓ With

platelet

agonists

CD, CD61

deficiency

Genetic testing may be

required if married within

family

Bernard Soulier

Syndrome N N N N N ND ND

↓ With

Ristocetin

CD 42b

deficiency

Genetic testing may be

required if married within

family

Storage pool

defects N N N N N ND ND

↓ With

some

agonists

ND

Platelet EM

studies,

Lumiaggre

gometry

Mild disorders

N- normal; ND- Not Done; NR- Not required; EM – Electron Microscopy

Page 373: Training Manual for Hemoglobinopathies and Hemophilia:

373

Management of Hemophilia and

other Bleeding Disorders

Page 374: Training Manual for Hemoglobinopathies and Hemophilia:

374

Clinical manifestations and management of hemophilia

Hemophilia is a bleeding disorder caused by a deficiency of coagulation factor VIII or IX.

Deficiency of factor VIII is known as hemophilia A and factor FIX is known as in hemophilia B.

The deficiency is due to mutations of the respective clotting factor genes resulting in factor

deficiency. Hemophilia A is more common accounts for 80 – 85 % of total hemophilia

population and the approximate incidence of HA is 1 in 10,000 births and HB is one in 25000 to

30000 births.

Hemophilia mainly affects males, women are carriers. In one third of patients there will not be

any family history of hemophilia and it is due spontaneous mutation of F8 and F9 genes.

Affected males will pass on the hemophilia gene to their daughters. Most carriers are

asymptomatic. Carriers with clotting factor levels of 40–60% of normal may sometimes have an

increased bleeding tendency. Female carriers can have moderate or severe deficiency of factors

due to extreme lyonization Female haemophilia can be due Turners syndrome or they may be

offsprings of female carriers and haemophilic fathers. Hemophilia is classified as mild,moderate

and severe depending on the factor level.

Table 14. Classification of Hemophilia based on severity and its relationship to bleeding

manifestations

Severity Factor level Clinical manifestations and

bleeding episodes

Severe < 1 IU/dL (< 0.01 IU/mL) or <

1% of Normal

Spontaneous bleeding into

joints or muscles,

predominantly in the

absence of identifiable

hemostatic challenge

Moderate 1–5 IU/dL (0.01 –0.05

IU/mL) or 1–5% of normal

Occasional spontaneous

bleeding and prolonged

Page 375: Training Manual for Hemoglobinopathies and Hemophilia:

375

bleeding with minor trauma

or surgery.

Mild

> 5–40 IU/dL (0.05–0.40

IU/mL) or > 5–40% of

normal

Severe bleeding with major

trauma or surgery.

Spontaneous bleeding is

rare.

Clinical manifestations

Clinically severe hemophilia A and B cannot differentiate unless there is family history and

accurate diagnosis is the first essential step in the management of hemophilia. A definitive

diagnosis depends on factor assays to demonstrate deficiency of FVIII or FIX. The severe

hemophilia patients usually present with joint or muscle bleeding and children may present later

when they start walking or running. Patients with mild haemophilia may not bleed excessively

until they experience trauma or surgery.

Hemophilia should be suspected in patients presenting with a history of:

Easy bruising in early childhood

Spontaneous” bleeding (bleeding for no apparent/known reason), particularly into the

joints, muscles, and soft tissues

Excessive bleeding following trauma or surgery

A family history of bleeding

Primary menorrhagia and postpartum bleeding

The hallmark of bleeding in patients with severe haemophilia is recurrent bleeding into muscles

and joints from an early age in life.

Hemarthrosis or joint bleed

Bleeding into joints in persons with hemophilia (PWH) leads to chemical reaction and synovial

hypertrophy, neovascularization which is prone for recurrent bleeding. It leads to cartilage

damage erosion and damage to subchondral bone. Hemophilic arthropathy most commonly

affects the large joints. The frequency of joint involvement in descending order: the knee, elbow,

ankle, hip, and shoulder .The ankles are most commonly affected in children.

Following are the ISTH SSC definitions of bleeding in hemophilia patients

Page 376: Training Manual for Hemoglobinopathies and Hemophilia:

376

An acute joint bleed : an unusual sensation (“aura”) in the joint (such as a feeling of tingling or

pressure) in combination with any of the following:

Increasing swelling or warmth of the skin over the joint

Increasing pain

Progressive loss of range of motion or difficulty in using the limb as compared with

baseline.

The progression of these symptoms over a relatively short period of time is an important feature

of an acute joint bleed (hemarthrosis).

How to differentiate between acute joint bleed and chronic hemophilic arthropathy? – Figure 3

It is clinically difficult to differentiate between two but the following can be useful

Rapid resolution of pain following infusion of factor concentrates - typical of an acute

haemarthrosis.

Improvement of pain associated with activity soon after a period of rest - typical of

chronic arthritis.

Reluctance to use the limbs alone - indicative of an acute joint/muscle bleed in infants

and young children.

Target joint

Three or more spontaneous bleeds into a single joint within a consecutive 6-month period.

Significance of target joint

It is prone to spontaneous bleeding that may be difficult to control with medical

measures such as prophylaxis

Target joints may need to be managed with synovectomy, either with radioactive

agents (preferable) or chemicals or removed arthroscopically, if possible.

Muscle bleed

An episode of bleeding into a muscle, determined clinically and/or by imaging studies generally

associated with pain and/or swelling and loss of movement over baseline.

Significance of muscle hemorrhage

Page 377: Training Manual for Hemoglobinopathies and Hemophilia:

377

Muscle bleed can occur in any of the muscles and early identification and proper management is

very important to prevent as it may lead to neurovascular compromise leading to permanent

contracture or pseudotumour.

Iliopsoas muscle hematoma : risk of femoro-cutaneous, cruralandfemoral nervepalsy

Superior-posterior and deep posterior compartments of the lower leg : risk of posterior tibial and

deep peroneal nerve injury

Flexor group of forearm muscles - risk of Volkmann’s ischemic contracture

PWH need to be monitored continuously for signs and symptoms of neurovascular compromise

or radiological investigations. In case of muscle bleed, it is difficult to quantify and can cause

significant blood loss; hence hemoglobin should be monitored. Iliopsoas hematoma patients

usually present with pain in the lower abdomen, groin or lower back, and pain on extension (but

not on rotation) of the hip joint. Some of them also present with paraesthesia on the lateral part

of the thigh or other signs of femoral nerve compression such as loss of patellar reflex and

quadriceps weakness. The symptoms may mimic acute appendicitis.As soon as the pain subsides,

physiotherapy should be started and some of the patients with iliopsoas hematoma may require

skin traction to straighten the limb which may needs be covered with factor infusion.

Re-bleed into a muscle/ joint: Bleeding occurring for more than 72 hours after stopping treatment

for the original bleed for which treatment was initiated.

Central nervous system haemorrhage or head trauma: After head trauma patients can develop

intracranial hemorrhage or forehead hematoma. It is a medical emergency and life threatening.

Patients present with headache, with or without vomiting, altered sensorium or they may become

unconscious. These are the patients who are prone to have recurrence and prophylaxis for 3 to

6months can be given. Neurosurgery consultation should be taken as early possible.

Tables 15 and 16 shows sites and frequency of bleeding in different sites.

Table 15:

Sites of bleeding in hemophilia

Severity Sites of bleeding

Serious Joints (hemarthrosis)

Muscles, especially deep compartments

(iliopsoas, calf and forearm)

Mucous membranes in the mouth, gums,

nose and genitourinary tract

Page 378: Training Manual for Hemoglobinopathies and Hemophilia:

378

Life-threatening Intracranial

Neck or throat

Gastrointestinal

Table 16:

Frequency of bleeding in different sites

Sites of bleeding Approximate frequency (%)

Hemarthrosis

more common into hinged joints (i.e.

ankles, knees and elbows)

less common into multiaxial joints (i.e.

shoulders, wrists and hips)

70–80

Muscle 10–20

Other major bleeds 5–10

Central nervous system 5

Principles of care

Primary aim of care is to prevent and treat bleeding with the deficient clotting factor.

Should be treated with specific factor concentrate in specific factor deficiency

Acute bleeds should be treated as quickly as possible, preferably within 2 hours.

Patients usually recognize early symptoms of bleeding even before the manifestation of

physical signs. This is often described as a tingling sensation or “aura”.

In life-threatening bleeding especially in the head, neck, chest, and gastrointestinal tract,

treatment with factor should be initiated immediately, even before diagnostic tests

All persons with hemophilia (PWH)should carry an identification card mentioning the

diagnosis, severity and contact information of treating physician or hospital.

Veins are the lifelines for PWH and should be handled carefully. 23 or 25G butterfly are

preferred

Drugs that affect platelet function, particularly aspirin and NSAIDs should be avoided.

Page 379: Training Manual for Hemoglobinopathies and Hemophilia:

379

Good oral hygiene is essential to prevent periodontal disease and dental caries, which

predispose to gum bleeding

Regular exercise and other measures to stimulate normal psychomotor development

should be encouraged to promote strong muscles, develop balance and coordination, and

improve fitness

Dosage of factor concentrates

World Federation of Hemophilia (WFH) strongly recommends the use of viral inactivated

plasma derived or recombinant factor concentrates in preference to fresh frozen plasma or

cryoprecipitate for haemophilia. Selection of clotting factor concentrates should be made on very

well laid down WFH guidelines on selection and tendering of clotting factor concentrates (CFC).

When selecting plasma-derived concentrates, consideration needs to be given to both the plasma

quality and the manufacturing process. Among the manufacturing process factors, purity of the

product and viral inactivation or elimination should be considered as the most important.

Types of treatment

Clotting factor concentrates can be infused on demand/ episodic or prophylactic or regular

replacement therapy. Episodic (on demand) replacement therapy is factor replacement therapy

given at the time of clinically evident bleeding. Prophylaxis is infusion of clotting factor

concentrates to prevent bleeding. Principle of prophylaxis is from the observation that moderate

hemophilia patients with clotting factor level > 1 IU/ mL seldom experience spontaneous

bleeding. Prophylaxis prevents and decreases bleeding and joint destruction and preserve normal

musculoskeletal function

Dosage of factor for bleed management:

Calculation of FVIII concentrates

Each unit of factor VIII infusion will raise the plasma FVIII level by about 2 IU/dl or by 2 % in

the body.

The dose is calculated as follows

Patients weight (in Kg) X (Desired level – Patient level) X 0.5

FVIII should be infused by slow intravenous injection at a rate not greater than 3 mL/minute in

adults or as specified in the product information leaflet.

Half-life of FVIII : 8 – 12 hours

Page 380: Training Manual for Hemoglobinopathies and Hemophilia:

380

If the PWH requires second dose or symptoms subside or in case of severe bleeding the next

dose should be half of the first dose after 8 to 12 hours or depending on the FVIII level.

Calculation of FIX concentrates

Each unit of factor IX infusion will raise the plasma FIX level by about 1 IU/dl or by 1 % in the

body.

The dose is calculated as follows

Patients weight (in Kg) X (Desired level – Patient level)

FIX should be infused by slow intravenous injection at a rate not greater than 3 mL/minute in

adults or as specified in the product information leaflet.

Half-life of FVIII : 18 – 24 hours

FIX concentrates should be infused by slow intravenous injection at a rate not greater than 3

ml/minute in adults, or as recommended in the product information leaflet.

Management of Target joint

Good management of PWH can prevent target joints. Once a target joint occurs (3 or more

bleeds in a single joint within 6 months), recurrent bleeding into this joint will continue at a

higher rate.Conservative management: Short-term prophylaxis for 4– 8 weeks can be used to

interrupt the bleeding cycle with intensive physiotherapy

Surgical treatment:Synoviorthesis either Chemical synovectomy Oxytetracycline or radioactive

synovectomy, or arthroscopic synovectomy

Emergency management in hemophilia

PWH should be triaged urgently and delay in treatment can significantly affect the

morbidity and mortality.

There should not be any delay in administering factors if PWH have any signs or

symptoms of bleeding and it should be given first without waiting for radiological

confirmation. If transport to another hospital is planned always give a dose of factor to

cover bleeding episode before transport. Early factor administration is life saving.

After investigations maintenance treatment can be planned.

Indications for Factor replacement therapy

Page 381: Training Manual for Hemoglobinopathies and Hemophilia:

381

Suspected bleeding into a joint or muscle.

Any significant injury to the head, neck, mouth or eyes or evidence of bleeding in these

areas.

Any new or unusual headache, particularly one following trauma.

Severe pain or swelling at any site.

All open wounds requiring surgical closure, wound adhesive or steri-strips.

History of an accident or trauma that might result in internal bleeding.

Any invasive procedure or surgery

Heavy or persistent bleeding from any site

Gastrointestinal bleeding.

Acute fractures, dislocations and sprains.

Any other signs or symptoms suggestive of bleeding

WFH recommendations for maintenance plasma factor level and duration in different sites of

bleeding in resource constraints situations is shown in Table 17.

Table 17.Suggested plasma factor peak level and duration of administration (from WFH 2020 guidelines).

This gives doses in both practise patterns of lower dose ( useful in resource constrained settings and higher

practise patterns. The dose can be modified based on experience, supply and patient factors. If uncertain,

consult a hemtologist with experience in these issues.

Page 382: Training Manual for Hemoglobinopathies and Hemophilia:

382

After replacement with factor concentrates the response can vary from excellent to none.

Assessment of bleeding response is essential and evaluation of next dose or inhibitor suspicion

will depend on response. Table 18 shows assessment of treatment of acute joint/muscle bleeds.

Table18.Assessment of treatment of acute joint/muscle bleeds

Category Response

Excellent Complete pain relief within 8 h and/or complete resolution of signs of

bleeding after the initial injection and not requiring any further

Page 383: Training Manual for Hemoglobinopathies and Hemophilia:

383

replacement therapy for relief of persistent symptoms and signs in

the same joint within 72 h.

Good Significant pain relief and/or improvement in signs of bleeding within

approximately 8 hours after a single injection, but requiring more

than one dose of replacement therapy within 72 h for complete

resolution.

Moderate Modest pain relief and/or improvement in signs of bleeding within

approximately 8 h after the initial injection and requiring more than

one injection within 72 h but without complete resolution.

None No or minimal improvement, or condition worsens within

approximately 8 hours after the initial injection

Physical activity

Physical activity should be promoted to improve physical fitness and normal

neuromuscular development and to concentrate on muscle strengthening, coordination,

general fitness, physical functioning, healthy body weight.

Contact sports such as cricket, tennis, football, basketball , boxing , wrestling etc should

be avoided

Non-contact sports such as swimming, walking table tennis and cycling under

observation

High contact and collision sports such as soccer, hockey, rugby, boxing, and wrestling,

motocross racing and skiing, should be avoided

Target joints can be protected with braces or splints during activity

Adjunctive treatment

For bleeding in muscles and joints

PRICE will relieve some symptoms and it is as follows

P – Protection with splints or compression bandages

R- Rest (new WFH 2020 guidelines have changed this to optimal loading (OL), meaning weight

bearing as soon as recovery occurs to prevent wasting from disuse of mucles. This changes the

acronym to POLICE)

I – Ice application

Page 384: Training Manual for Hemoglobinopathies and Hemophilia:

384

Ice or cold packs may be applied around the joint for 10–20 minutes every 2–4 hours for

pain relief.

Ice should never be applied in direct contact with skin.

Crushed ice wrapped in towel can be applied all over the joint

C-Compression

E-Elevation of the joint

Rehabilitation - Regular exercise is also the most important part in the management of PWH. As

soon as the pain and swelling begin to subside the patient should be encouraged to do isometric

exercises and then isotonic exercises with complete extension. Early active muscle control

mustbe encouraged, to minimise muscle atrophy and prevent chronic loss of joint motion

Antifibrinolytic drugs

Tranexamic acid is an antifibrinolytic agent that competitively inhibits the activation of

plasminogen to plasmin and it promoted clot stability. Oral Tranexamic acid tablets, 25 mg/Kg

or up to 1.5 g in adults, three to four times daily. Tranexamic acid is effective as adjunctive

treatment for mucosal bleeds and dental extractions. The use of tranexamic acid is

contraindicated for the treatment of haematuria because its use may prevent dissolution of clots

in the ureters, leading to serious obstructive uropathy and potential permanent loss of renal

function.

Epsilon amino caproic acid can also be used epsilon aminocaproic acid (EACA) 50 to

100 mg/kg every 6 hours in children and in adults loading dose of 4 to 5gm followed by lower

doses every 6 to 12 hourly(total suggested dose is 100 mg/kg/d).

Vaccinations

All bleeding disorders patients should be completely vaccinated including hepatitis A and

B

All the vaccines should be given subcutaneously

Intramuscular injections should be avoided.

Tetanus and Pertussis vaccine needs to be given intramuscularly

Infusion factor concentrates along with vaccine has not shown to increase inhibitor

chances however it is better to avoid CFC infusion prior to vaccination

Page 385: Training Manual for Hemoglobinopathies and Hemophilia:

385

Smallest gauge needle available (usually 25–27 G) should be used

An ice pack can be applied to the injection site for 5 mins and to apply pressure to the

injection site for at least 5 to 10 mins

Live virus vaccines (such as oral polio vaccine, MMR) may be contraindicated in those

with HIV infection

Pain management

Acute and chronic pain are common in patients with haemophilia. Adequate assessment of the

cause of pain is essential to guide proper management.

Pain caused by venous access : Local anaesthetic spray or cream can be used for paediatric

patients.

Pain caused by joint or muscle bleeding : Clotting factor concentrates should be infused as early

as possible to stop bleeding, other measures such as ice packs, splints crutches and pain

medication should be used

Postoperative pain: Intramuscular injection of analgesia should be avoided and pain strategy

should be followed.

Pain due to chronic hemophilic arthropathy: Functional training, adaptations and adequate

analgesia.

Table 19 .Strategies for pain management in patients with haemophilia.

No. Strategy

1 Paracetamol or acetaminophen If not effective, then use Strategy 2

2 COX-2 inhibitor (e.g. celecoxib, meloxicam and others) OR Paracetamol or

acetaminophen plus tramadol (3–4 times/day). If not effective, use Strategy 3

3 Opiate analgesia: use a slow-release product such as controlled-release

oxycodone, tramadol SR or controlled-release morphine

Dental care and management

Teeth should be brushed twice a day with a medium-texture brush to remove plaque

deposits

Good oral hygiene is essential to prevent periodontal disease and dental caries which may

predispose to gum bleeding

Page 386: Training Manual for Hemoglobinopathies and Hemophilia:

386

Dental examinations should be conducted regularly in all PWH

Dental floss or interdental brushes should be used wherever possible

Toothpaste containing fluoride should be used in places where fluoride content is not

present in water supply.

Toothpaste containing fluoride should be used in areas where natural fluoride is not

present in the water supply. Fluoride supplements may also be prescribed if appropriate.

Close liaison between the dental surgeon and the hemophilia team is important for good

dental care

Tranexamic acid or EACA is often used after dental procedures to reduce the need for

replacement therapy

Oral antibiotics should only be prescribed if clinically necessary.

Local hemostatic measures like fibrin glue or local tranexamic acid may also be used

Following a tooth extraction, the patient should be advised to avoid hot food and drinks

until normal feeling has returned.

Regular warm salt water mouthwashes (a teaspoon of salt in a glass of warm water)

should begin the day after treatment and continue for 5–7 days or until the mouth has

healed.

Prolonged bleeding and/or difficulty in speaking, swallowing, or breathing following

dental manipulation should be reported to treating hemophilia physician and dental

surgeon

Venous access

Veins must be treated with care

They are the lifelines for a person with haemophilia.

23- or 25-gauge butterfly needles are recommended.

Apply pressure for 3–5 min after venipuncture.

Venous access devices should be avoided whenever possible, but may be required in

some children.

Comprehensive care centre for Hemoglobinopathies & Haemophilia

Comprehensive care centre promotes physical and psychosocial health and quality of life while

decreasing morbidity and mortality.Hemophilia care is beyond just factor replacement and

management includes improvement in health and quality of life of patient and his family. The

wide-ranging needs of people with hemophilia and their families are best met through the

Page 387: Training Manual for Hemoglobinopathies and Hemophilia:

387

coordinated delivery of comprehensive care by a multidisciplinary team of healthcare

professionals.

The team consists of

Medical director (preferably a pediatric and/ or adult hematologist or a physician with

interest and expertise in hemostasis)

Nurse coordinator who coordinates the provision of care , educates patients and their

families ; acts as the first contact for patients with an acute problem or who require

follow-up and is able to assess patients and institute initial care where appropriate

Musculoskeletal expert (physiotherapist, occupational therapist, physiatrist, orthopaedics

surgeon, rheumatologist) who can address prevention as well as treatment.Experience

with PWH is preferred, or training can be undertaken.

A Hematologylaboratory specialist.

A psychosocial expert (preferably a social worker, or a psychologist) familiar with

available community resources

Members of the hemophilia care team: The comprehensive care team should also include or have

access to chronic pain specialist,dentist, geneticist, hepatologist, infectious disease specialist ,

immunologist, gynecologist/obstetrician and vocational counsellor etc.

Functions of a comprehensive care program

Provide or coordinate inpatient (i.e., during hospital stays) and outpatient (clinic and other

visits) care and services to patients and their family.

Patients should be seen by all core team members at least yearly (children every 6 months)

for a complete hematologic, musculoskeletal, and psychosocial assessment and to develop,

audit and refine an individual’s comprehensive management plan.

The plan of further management should be made and should be communicated to all

treaters and care facilities.

Communication among treaters

Advice to smaller centers and personal physicians and provide primary care and

management of some complications in frequent consultation with the comprehensive care

centre (particularly for patients who live a long distance from the nearest hemophilia

treatment centre).

Page 388: Training Manual for Hemoglobinopathies and Hemophilia:

388

Train and supervise home therapy where ever feasible

Educate patients, family members and other caregivers to ensure that the needs of the

patient are met

Collect data on sites of bleeds, types and doses of treatment given, assessment of long-term

outcomes (particularly with reference to musculoskeletal function), complications from

treatment, and surgical procedures. All this information should be recorded in registry.

Home therapy

Home therapy improves adherence to prophylaxis, allows immediate access to clotting factor

with early treatment resulting in decreased pain dysfunction and long-term disability improving

the quality of life, freedom to travel, less absenteeism and greater stability

What is the importance of Inhibitors?

Inhibitors are alloantibodies to factor VIII or IX and is an important complication of treatment.

Inhibitor testing should be done in a PWH who has inadequate response to optimum factor

correction and regularly in patients on prophylaxis which will be discussed in the next training

module. Neutralize the function of infused factor VIII or IX clotting factor concentrates, that is

the factor concentrates become ineffective to control bleeding.

Clinically relevant - Inhibitors should be documented on two separate occasions within a 1- to 4-

week period and should have a level of ≥ 0.6 Bethesda Units (BU/mL) using the Nijmegen

modification of the Bethesda assay. Inhibitors are discussed in the following section of training

module.

Documentation of treatment and data monitoring is the most important aspect in the management

of PWH. At least the annual bleed rate and school or parent absenteeism should be documented

to access the efficacy of any treatment. Following Joint assessment and functional activity

should be monitored at least on an annual basis

Clinical: WFH Physical Examination Score (aka Gilbert score), Hemophilia Joint Health

Score (HJHS)

Radiological: Pettersson score, MRI, and ultrasound scores

Activity: Hemophilia Activities List (HAL), Paediatric Hemophilia Activities List

(PedHAL), Functional Independence Score in Hemophilia (FISH)

Page 389: Training Manual for Hemoglobinopathies and Hemophilia:

389

Health-related quality of life: (HaemoQol, Canadian Hemophilia Outcomes: Kids’ Life

Assessment Tool [CHO-KLAT])

Figure 11.Images of different sites of hemarthroses and hematomas in patients with

Haemophilia

A. Patient with hemophilia with left elbow acute hemarthrosis

B. Child with hemophilia with forehead hematoma after trauma

C. Patient with hemophilia with left knee chronic hemarthrosis

Suggested Reading

Srivastava, A, Santagostino, E, Dougall, A, et al. WFH Guidelines for the Management of

Hemophilia, 3rd edition. Haemophilia. 2020: 26(Suppl 6): 1‐ 158. https://doi.org/10.1111/hae.14046

Srivastava, A. K. Brewer, E. P. Mauser‐ Bunschoten et al. Guidelines for the management

of hemophilia. Haemophilia 2013: 19; e1-e. 47.

Victor S. Blanchette, Alok Srivastava. Definitions in Hemophilia: Resolved and Unresolved

Issues. Semin ThrombHemost 2015; 41:819–825.

Guidelines for emergency department management of individuals with hemophilia and other

bleeding disorders. MASAC Document #252 Replaces Doc #175

J. Hanley, a. Mckernan,† m. D. Creagh et al. Guidelines for the management of acute joint

bleeds and chronic synovitis in haemophilia A United Kingdom Haemophilia Centre

Doctors’ Organisation (UKHCDO) guideline. Haemophilia 2017; 1–10.

Guidelines for the management of haemophilia in Australia. Haemophilia Guidelines July

2016. National Blood Authority. Locked Bag 8430. Canberra ACT 2601

Page 390: Training Manual for Hemoglobinopathies and Hemophilia:

390

Management of hemophilia with inhibitors

Inhibitor development is a serious complication which occurs in some patients with hemophilia.

Approximately 33% patients with severe hemophilia A will develop inhibitors, some inhibitors

are transient but at other times they may persist.

When inhibitors occur, the factor replacement treatment becomes ineffective and risk

of life threatening hemorrhages again appear; these recurrent bleeds compromise joint

health and quality of life

At present in severe hemophilia A, only prophylaxis has been shown to possibly reduce the risk

of inhibitor development.

Some important factors which increase the risk of development of inhibitors are

Family history (siblings, cousins, uncles etc)

High risk mutations: null mutations, inversions,large deletions and mutations inducing

stop codons.

Intense exposure- High dose factor replacement during surgery

Infection

Early exposures, especially first 50 exposure days (ED) with a median time of inhibitor

presentation at about 10-15 EDs.

Hemophilia A: Age at first exposure does not appear to have an effect, though practically risk is

highest below the age of 5 years. There is an increased risk after the age of 60 years.

Haemophilia B: FIX inhibitors are associated with serious allergic reactions to FIX, especially in

those with gene deletions. Any reaction should prompt inhibitor testing before further FIX

exposure as even low-level FIX inhibitors may cause anaphylaxis

Page 391: Training Manual for Hemoglobinopathies and Hemophilia:

391

When to suspect inhibitors ?

Inhibitor testing should be performed if a patient has a poor clinical response to

concentrate or lower FVIII/IX levels than expected after concentrate infusion.

• Unsuspected increase in number or severity of bleeding episodes,

• Lesser response to previously used factor concentrates or less than expected recovery

(FVIII levels)

• Unusual bleeding episodes i.e. muco-cutaneous, GI, urogenital etc

When to measure inhibitors?

An inhibitor screen should be performed in patients with severe hemophilia frequently

till 30 exposures- recommended every fifth dose.

Thereafter, inhibitor testing should be undertaken every 3–6 months until 150 EDs.

Then annually or with increasing bleeding episodes.

How to measure inhibitors?

• The Specific assay - Bethesda assay (BA) or if available Nijmegen modified Bethesda assay

(NBA) should be done. The Inhibitor assay is positive when the titer is >0.6BU/mL.

If Bethesda assay/ Nijmegen modified Bethesda assay for inhibitors not available,

then an incubated mixing assay can be done.

If positive- then refer patient for inhibitor testing as soon as possible as this is a

serious problem.

Do not continue factor replacement till results are obtained.

More about inhibitors

• Titers of inhibitors: A low responding inhibitor is defined as an inhibitor level that is persistently

< 5 BU/mL, whereas a high responding inhibitor is defined by a level ≥ 5 BU/mL.

• High responding inhibitors tend to be persistent. If not treated for a long period, titer levels may

fall or even become undetectable, but there will be a recurrent anamnestic response in three to

five days when challenged again with anti - hemophilic factor products

• Some low titer inhibitors may be transient, disappearing within six months despite antigenic

challenge with factor concentrate.

Page 392: Training Manual for Hemoglobinopathies and Hemophilia:

392

· Very low titer inhibitors may not be detected by the Bethesda inhibitor assay, but by a poor

recovery and/or shortened half-life (T-1/2) following clotting factor infusions.

How to manage hemophilia patients with inhibitors?

The management / treatment of haemophilia patients with inhibitors has two components:

Treatment or prevention of acute bleeding

Eradication of inhibitors by Immune Tolerance Induction (ITI)

Treatment of acute bleed

If the inhibitor titre is low and low responding (inhibitortiter does not increase with factor

challenge, and historical inhibitor peak levels never exceeded 5 BU/mL), such cases can treated

with higher doses of factor VIII.

If this information not available or high titre inhibitor, then treat bleeds with bypassing

agents only.

Refer patient to Level 3 or Level 4 for complete laboratory evaluation and clinical

management.

High titer inhibitors: If high-titer, high-responding inhibitors (historical inhibitor peak levels that

exceeded 5 BU/mL at least once) are present, then FVIII/FIX concentrates are contra indicated.

Currently two treatment modalities are approved for this indication:

1. Bypassing Agents: aPCC (FEIBA®), rFVIIa (NovoSeven®)

2. Bispecific Monoclonal Antibody: Emicizumab.

Currently available and commonly used Bypassing agents:

Two by-passing agents are commercially available plasma-derived aPCC (FEIBA®), and rFVIIa

(NovoSeven®).

Both are clinically effective, however individual variations may occur, if the patient does not

respond to appropriate dose of one agent then the alternative agent is to be tried. Reasons for this

individual variation, is not known. Give BPA dose to stabilize and refer to higher center.

Page 393: Training Manual for Hemoglobinopathies and Hemophilia:

393

Table 20.

Characteristics of Bypassing agents

rFVIIa (NovoSeven®) aPCC (FEIBA®)

Mechanism of action

Half-life

Mode of Administration

Dosing and frequency

Acute Bleeding

Prophylaxis

Activates FX on the surface of

platelets

2-3 hours

IV

90-120µg/kg repeat doses

every 2-3 hourly

Maybe used three times a

week

Action of FXa and FII

6-12 hours

IV

50-100 U/kg 2-3 times daily [Max.

dose is 200 U/kg/day]

More evidence to support use.

Dose is usually

85 units /kg three times/week

Development of inhibitors leads to increased mortality and poor quality of life. Patient should be

seen in a center of excellence or tertiary care center (Level 3 or 4), The recommendations from

the center should be followed.

The standard Bypassing agents have been FEIBA and Recombinant factor 7a (rFVIIa)

• 2 doses of rFVIIais equivalent to one dose FEIBA in managing a joint bleed.

• Some patients respond better to one bypassing agent- switch if no response

rFVIIa:

• rFVIIa Dose 90mcg/kg iv. Every 2 -3 hours,

• Better response with 270mcg/kg single dose

FEIBA/APCC:

FEIBA dose 50-100 units/kg/day to manage bleeding,

• Max dose 200 units/kg/day, can be repeated every 6-12 hours.

• Risk of anamnestic increase in inhibitor (but more in FIX patients)

Page 394: Training Manual for Hemoglobinopathies and Hemophilia:

394

• To prevent bleeding in inhibitor patients FEIBA can be given on alternate days to prevent

bleeding, this is prophylactic use of bypassing agents. This is better than only treating

bleeds as study has shown better joint outcome.

Newer agents: Emicizumab is recombinant humanized bispecific monoclonal antibody that

bridges activated factor IX and factor X to replace the function of missing activated factor VIII,

thereby restoring hemostasis. This can be an effective agent for haemophilia patients with

inhibitors.Emicizumab is recombinant humanized bispecific monoclonal antibody that bridges

activated factor IX and factor X to replace the function of missing activated factor VIII, thereby

restoring hemostasis. The antibodies against FVIII do not bind or neutralize Emicizumab and it

bypasses the need for factor VIII hence, this is clinically effective in all individuals with

haemophilia A- those with inhibitors or those without inhibitors. Clinical trials suggest Ctrough

≥45 μg/Ml should result in a median ABR of 0 and Emicizumab is approved only for

Prophylactic treatment.

Table 21.Characteristics and dose of Emicizumab (Hemlibra)

EMICIZUMAB

Mechanism of action

Half-life

Mode of

Administration

Dosing and frequency

Prophylaxis

Replaces the action of FVIIIa by binding with both FIXa

and FX to trigger the activation of the rest of the

coagulation cascade

4 Weeks

SC (subcutaneous injection)

3mg/kg for 4 weeks loading Dose (to attain Ctrough ≥45

μg/Ml)

1.5 mg/ kg/ week or 3mg/kg once in 2 weeks

maintenance dose

Page 395: Training Manual for Hemoglobinopathies and Hemophilia:

395

Immune tolerance induction ( ITI)

Immune tolerance induction (ITI) is at present the only treatment which can eradicate

inhibitors. ITI consists of regular, frequent, and prolonged exposure of the patient to specific

factor concentrates to induce tolerance. Immune tolerance is reported to be highly effective and

about 70% of patients become inhibitor free. This therapy should only be performed at level 3/ 4

centers. Interruption of ITI, infections, and patients with high titers of inhibitors or long history

of persistent inhibitors have less chance of success. Success is considered when inhibitor titer

can no longer be measured, factor recovery is greater than 66% of normal and half-life of Factor

VIII is greater than 6 hours.

In ITI, regular replacement of factor is given at higher doses, with monitoring and management

of breakthrough bleeds with bypassing agents till the inhibitor disappears. This process may take

months to years. After eradication of inhibitors the patients need to be put on prophylaxis to

decrease recurrence of inhibitors in the patient.

There are several regimens for ITI, two protocols can be followed for ITI in consultation with

Level 3 or 4 center with experience with ITI treatment.

Bonn Protocol :

Hemophilia. A: FVIII 200-300 IU/kg daily + FEIBA 100 units/kg twice daily and Van

Crevald (Dutch or low dose): FVIII 25 -50 IU/kg on alternate days and escalated. Currently

many HTC use Factor VIII 50 units /kg three times a week and monitor. The higher dose ITI in

the International ITI study was shown to have less bleeding episodes.ITI therapy will take

6months or longer the remove the inhibitor. After ITI patients have to be put on prophylaxis, to

prevent recurrence. About 20 -30% patients may fail, then a different ITI regimen may be tried

or immunosuppressive/ immunomodulation therapy has been advocated, after failure of

conventional ITI by WFH.

Hemophilia B: Experience with ITI for hemophilia B inhibitor patients is limited. The principles

of treatment in these patients are similar, but the success rate is much lower, especially in

persons whose inhibitor is associated with an allergic diathesis. Hemophilia B inhibitor patients

with a history of severe allergic reactions to FIX may develop nephrotic syndrome during ITI,

which is not always reversible upon cessation of ITI therapy. Alternative treatment schedules,

including immunosuppressive therapies, are reported to be successful.

Page 396: Training Manual for Hemoglobinopathies and Hemophilia:

396

Suggested reading

Srivastava, A, Santagostino, E, Dougall, A, et al. WFH Guidelines for the Management of

Hemophilia, 3rd edition. Haemophilia. 2020: 26(Suppl

6): 1‐ 158. https://doi.org/10.1111/hae.14046

Charles R. M. Hay and Donna M. DiMichele, on behalf of the International Immune Tolerance

Study. The principal results of the International Immune Tolerance Study: a randomized dose

comparison. Blood 2012; 119:1335-1344.

Minno GD, Santagostino E, Pratt K, Königs C.

New predictive approaches for ITI treatment.Haemophilia. 2014 Sep;20 Suppl 6:27-43.

Seth, Tulika. (2019). Experience of Immune Tolerance Induction Therapy for Hemophilia A

Patients with Inhibitors from a Single Center in India. Indian Journal of Hematology and Blood

Transfusion. 36. 10.1007/s12288-019-01218-2

Implementation of Prophylaxis for Persons with Hemophilia (PWH) in

India

1. What is prophylaxis?

Prophylaxis is the regular infusion of clotting factor concentrates in order to prevent bleeding.

The idea of prophylaxis came from the observation that people with moderate or mild

haemophilia (who have clotting factor levels of 1% or more) rarely experience spontaneous

bleeding. They also have less joint damage than people who have severe haemophilia.

2. What are the different types of prophylaxis?

Page 397: Training Manual for Hemoglobinopathies and Hemophilia:

397

Unlike episodic or “on demand” treatment, which is given at the time of a bleed to make it stop,

prophylaxis is given to prevent bleeding before it starts.

There are several types of prophylaxis. Continuous prophylaxis (primary, secondary, and

tertiary) is given regularly over a period of several months and often years. Intermittent or

periodic prophylaxis is given for shorter periods of time, usually a few weeks or months.

Table 22. Definitions of types of therapy for hemophilia

Type of Treatment Definition

Episodic (“on demand”) treatment Treatment given at the time of bleeding.

Continuous prophylaxis

Primary prophylaxis

Regular continuous treatment, started before the

second large joint bleed and age of 3 years.

Secondary prophylaxis

Regular continuous treatment started after 2 or

more large joint bleeds but before the onset of

joint disease.

Tertiary prophylaxis Regular continuous treatment started after the

onset of joint disease to prevent further damage.

Intermittent (“periodic”) prophylaxis

Treatment given to prevent bleeding for short

periods of time, such as during and after surgery

(not exceeding 45 weeks in a year)

Adapted from: Guidelines for the Management of Hemophilia, World Federation of Hemophilia, 2012.

Why prophylaxis is the way forward in the management of PWH in India?

The treating Physicians believed that if they could keep minimum factor levels around 1% with

regular infusions of clotting factor concentrates, they may reduce the risk of bleeding and

prevent joint damage. Since then, important studies have shown that children who receive

prophylaxis do have fewer bleeds and healthier joints. There is a false notion that prophylaxis is

more expensive than “on demand” therapy but studies have clearly shown that low dose

prophylaxis leads to less factor consumption and is more cost-effective than treating the bleeds

Page 398: Training Manual for Hemoglobinopathies and Hemophilia:

398

after they occur (ie. “on demand”). Prophylaxis is now the goal of treatment for people with

severe haemophilia, allowing them to remain active and participate more fully in daily life.

Caveat

Prophylaxis will not help repair joints that are already damaged. However, it will decrease the

frequency of bleeding, may slow progression of joint disease, and may improve quality of life.

Checklist before starting prophylaxis in your hospital

Are there motivated children and parents who would regularly visit your

hospital to take the injections? (unless carrying of factors to home and

storage for home therapy is permitted by local law)

Have you calculated the weekly requirement of total factors for the

group?

Have you ensured that the stock is adequate so that continuous

prophylaxis can be provided?

Have you sensitized the PWH that unless they perform regular exercise

the joint health will not improve?

Have you put in place a system for periodic screening for inhibitors as

per standard guidelines?

Are the monitoring charts for periodic assessment of joints and

functional activity ready?

Factors to consider when designing prophylaxis protocol

Age at which prophylaxis started (the earlier started, the better)

Current age (may be difficult to continue throughout life)

Venous access (butterfly needles with minimum damage to the veins)

Bleeding symptoms (frequent bleeders could be more benefitted)

Joint status (joints should not be damaged to such an extent that there is

Page 399: Training Manual for Hemoglobinopathies and Hemophilia:

399

Factors to consider when designing prophylaxis protocol

no reversible component)

Level and timing of physical activity (proper increment of factor levels

should be documented especially if the bleeding rate does not decrease)

Availability of clotting factor concentrates (uninterrupted supply is a

must)

Size(s) of vials of clotting factor concentrates available (larger dose vials

may be shared between multiple children to prevent wastage)

3. How to select the schedule and administer prophylaxis at your hospital ?

Studies are still underway to determine the best dosing schedule (also called a ‘protocol’). A

prophylaxis schedule should clearly outline:

The type of factor product to be used (Long acting factor concentrates are not available in

our country at this point of time)

The dose of factor administered with each injection

The frequency at which treatment is administered

The time (of day, or week) that treatment is administered

There are currently two protocols in use for which there is long-term data:

The Malmö protocol: Injections of 25-40 IU/kg, administered three times a week for those with

haemophilia A and twice a week for those with hemophilia B.

The Utrecht protocol: Injections of 15-30 IU/kg, administered three times a week for those with

haemophilia A and twice a week for those with hemophilia B.

However, these protocols are not practically feasible in most parts of the world including India

In countries with significant resource constraints like India, lower doses of prophylaxis given

more frequently (e.g., 10-15 IU/kg, 3 times per week) may be an effective option.[2-5]. The long

term benefits of these low dose prophylaxis regimens is not yet established and data should be

collected if using these newer regimens or it should be done under a clinical trial.Prophylaxis

should be administered to children to make it cost effective and ensure better outcomes.

In recent studies, it has even been proven that a low dose protocol as detailed below can

significantly reduce the annual bleeding rate and prevent joint damage. This kind of treatment

Page 400: Training Manual for Hemoglobinopathies and Hemophilia:

400

regimen is also associated with better school attendance and reduced hospitalization among

children with hemophilia.

Factor VIII concentrate may be given at a dose of 20-40 IU/kg in two divided doses per week for

hemophilia A (ie 10-20 IU/kg per dose).[6]

The dosage of factor IX concentrate for hemophilia B is 25- 40 IU/kg/wk.

The dosage is to be adjusted to the nearest available vial dose at the time of visit.

The frequency of administration can be increased to thrice a week for children with Haemophilia

A and twice a week for children with Hemophilia B if they have excessive bleeding.

Screening for inhibitors is an integral part of this management protocol. Inhibitor development is

particularly common in people with severe hemophilia within the first 75 treatments with

clotting factor concentrates. More than 50% of inhibitors develop within the first 15 exposure

days.

4. How do you assess the efficacy of the prophylaxis regimen?

More than three decades of research has shown that continuous prophylaxis is preferable to on-

demand therapy to reduce the frequency of bleeding and to prevent or delay joint damage.

People with haemophilia who are receiving prophylaxis should have an assessment on a regular

basis to ensure that the goals of therapy are being met and to make any necessary adjustments to

the treatment plan. These assessments should include an evaluation of:

Joint health/status

Bleed frequency

Limitations in activities

Psychosocial integration

5. What are the barriers and challenges to implementing prophylaxis in India?

Cost and access to treatment products

The biggest barrier to long-term prophylaxis is the cost of treatment. Prophylaxis is only possible

if significant resources are allocated to hemophilia care. However, it is cost-effective in the long-

term because it eliminates the high cost associated with subsequent management of damaged

joints and improves quality of life.

Page 401: Training Manual for Hemoglobinopathies and Hemophilia:

401

With NHM allocating significant resources for hemophilia care in India, prophylaxis is now a

reality for children with Hemophilia in India. It is very important to collect and provide scientific

evidence that justifies the high cost of treatment. Follow-up data of these patients will be very

important in confirming the effects on their joints. Cost-efficacy studies designed to identify

minimum dosage are necessary to allow access to prophylaxis across the country.

Venous access

Prophylaxis requires frequent injections and it can be difficult to find suitable veins in very

young children with hemophilia. In our country, all children can be administered early

prophylaxis without venous access devices provided good care of veins is taken.

Adherence

Adherence to (or compliance with) a treatment plan is generally defined as the extent to which

patients take medications as prescribed by their healthcare providers. According to the World

Health Organization, rates of non-adherence with any medication treatment may vary from 15%

to 93%, with an average estimated rate of 50%.

Adherence to a prophylaxis protocol is critical to its success. Prophylaxis is most effective if

factor levels are continuously maintained above the target level. Missing or skipping a dose can

cause clotting factor levels to fall below this target, which increases the risk of bleeding.

Bleeding that occurs while a patient is on prophylaxis is called ‘breakthrough bleeding’.

Patients and healthcare providers must work together to ensure that the protocol is manageable

for the person with haemophilia and their family. A patient’s adherence to the protocol should be

assessed regularly during clinic visits and strategies to improve adherence, including changes to

the protocol, should be explored wherever possible.

Suggested reading

Srivastava A, Brewer AK, Mauser-Bunschoten EP, Key NS, Kitchen S et al. Guidelines for

the management of hemophilia. Haemophilia, 2013; 19: p. e1-47.

Verma SP, Dutta TK, Mahadevan S, Nalini P, Basu D, Biswal N, et al. A randomized

study of very low-dose factor VIII prophylaxis in severe haemophilia - A success story from

a resource limited country. Haemophilia : the official journal of the World Federation of

Hemophilia. Haemophilia 2016; 22: 342-8.

Page 402: Training Manual for Hemoglobinopathies and Hemophilia:

402

Poon, .C and Lee A. Individualized prophylaxis for optimizing hemophilia care: can we

apply this to both developed and developing nations? Thrombosis Journal, 2016; 14: 32.

Dunkley S, Lam JCM, John MJ, Wong RSM, Tran H, Yang R, et al. Principles of

haemophilia care: The Asia-Pacific perspective. Haemophilia 2018; 24: 366-75.

Carcao M. and Srivastava A. Factor VIII/factor IX prophylaxis for severe hemophilia.

Seminars in Hematology 2016; 53: 3-9.

Sidharthan NR. Sudevan V. Narayana Pillai S. Mathew M. Raj D. Viswam C.et al. Low-

dose prophylaxis for children with haemophilia in a resource-limited setting in south India-A

clinical audit report. Haemophilia2017; 23: e382-e384.

Surgery in hemophilia

Surgery in hemophilia requires special care since the underlying problem of patients with

haemophilia is defective blood clotting. Only specialised centres should attempt surgical

procedures since a well equipped laboratory is a must.

What is the aim?

The goal is to achieve satisfactory hemostasis during the procedure as well as perioperative

period so that a stable blood clot is formed which will lead to wound healing.

Is it required to achieve normal level of factors for surgery?

Even though normal level of factors range from 50-150%, it is not required to achieve 100% for

effective hemostasis during surgery or during perioperative period.

Maximum levels are required during the first three days of surgery.

Once a stable blood clot forms by this time, lower levels are sufficient.

Page 403: Training Manual for Hemoglobinopathies and Hemophilia:

403

This in effect means higher doses for the first three days and lower doses subsequently.

What are the surgical procedures possible?

All surgeries done in a patient with normal haemostatic system can be done in a patient with

hemophilia. However the level of factor support required varies depending upon the type and

severity of hemophilia and the type of surgery planned.

How are surgeries classified as minor and major in patients with Hemophilia?

Although there are no strict definations, the level of surgical invasiveness is most

commonly used as a defining factor for major surgery, with criteria including: a

requirement for a general or spinal anesthetic, provision of respiratory assistance,

abdominal surgery, penetration of a major body cavity, orthopedic surgery (joints), and

extraction of more than 3 teeth .

World Federation of Hemophilia classifies “A major surgical procedure is defined as

one that requires hemostatic support for periods exceeding 5 consecutive days”.

Table 23. Classification of types of Surgery for Hemophilia

General Surgery Orthopedic Surgery

Major “-ectomy” procedures

“-otomy” procedures

Pseudotumor

Dental -Extraction >3 teeth

Osteotomy/arthrodesis Dentala

Joint replacement/ arthroplasty

Synovectomy/arthroscopic synovectomy

Bone fracture reduction

Osteosynthesis,Arthroscopy,Amputation

Minor Centralline insertion/removal

Liver biopsy

Prostate biopsy, colonoscopy

with biopsy and other

diagnostic biopsies

Abcess-drainage,

Wound suturing,circumcision

Chemical-/radio-/arthroscopic synovectomy

Mild (factor level 5-30%) and moderate (factor level 1-5%) hemophilia generally require

lower level of factor support compared to severe hemophilia (factor level <1%).

Hemophilia B requires lower level than hemophilia A

Page 404: Training Manual for Hemoglobinopathies and Hemophilia:

404

How long should factor support be given?

Generally factor support is required till wound healing or suture removal whichever is earlier.

This approximately corresponds to 3 days for minor procedures and 10 days for major

procedures. This can be longer depending on the particular type of surgery performed.

What are the pre requirements?

Additional requirements over and above in the usual setting

Laboratory should have the facility to do the following.

Factor assay

Bethesda assay

PT, APTT, fibrinogen if required post operatively

Ideally, full diagnostic testing of coagulation disorders

Clinical care should have the following human resources.

Hematologist

If not, a Physician well trained in managing coagulation disorders including support

during and after surgery

Ideally, Surgeons familiar with handling of patients with disorders of coagulation

How much factor support is required?

This will depend on the level and duration of factor support planned. Even though higher levels

are followed in countries without resource constraints, it has been shown that lower levels will

work equally well from studies done in developing countries, including India. Approximately

300 IU/kg of factor support will be required for a major procedure without complications.

How much blood and blood product support is required?

Like all surgical procedures, hemophilia patients will also require blood and blood product

support. If planned levels of factor are achieved, higher than anticipated blood loss is unlikely to

occur.

Is there any other support which will help?

Antifibrinolytic agents like tranexamic acid aid in forming a stable clot and should be routinely used.

However they are contraindicated in urological procedures.

Page 405: Training Manual for Hemoglobinopathies and Hemophilia:

405

What all things should be done prior to the planned surgery?

Correct diagnosis of the coagulation disorder, especially whether it is haemophilia A or

B.

If it is mild or moderate haemophilia, rule out von Willebrand disease.

Bethesda assay to look for inhibitor status, if high titre, factor will be ineffective and

patient will require bypassing agents like FEIBA or rVIIa.

If low titre, should look for recovery assay and for rise in titre 3-5 days following factor

infusion.

How do we know it is safe to do surgery after giving factors?

Factor levels should be checked after giving factor prior to the surgery, this is called a

recovery assay.

Subsequently it should be checked on days 1,3,5 and 7 of post operative period, these are

the trough levels.

Dose should be modified if satisfactory levels are not achieved or if higher than planned

levels are achieved.

Table 24.Recommended factor support for resource constrained settings

Day Target level Factor dose

Hemophilia A Hemophilia B Hemophilia A Hemophilia B

0 (Recovery) 80-100% 60-80% 40-50 IU/kg Q12H 60-80 IU/Kg OD

1-3 (Trough) 40-50% 30-40% 20-25 IU/kg Q12H 30-40 IU/kg OD

3-5(Trough) 30-40% 20-30% 15-20 IU/kg Q12H 20-30 IU/Kg OD

>5(Trough) 20-30% 10-20% 10-15 IU/kg Q12H 10-20 IU/kg OD

How are the peak levels and trough levels defined and how should they be monitored ?

Peak Level :- The peak factor level is the maximum factor level measured within 1 hour

of a bolus injection. Wherever feasible, peak level should be measured immediate pre op and

Page 406: Training Manual for Hemoglobinopathies and Hemophilia:

406

should be targeted to 60-80U/dl at the minimum and can be kept upto 100% in neurologic

surgeries.

Trough Level :- The trough factor level is the minimum factor level measured

immediately before the next bolus injection.. Wherever feasible the dosage in patients

undergoing major surgery should be titrated as per the trough which should be kept between 30-

50IU/dl for initial 2 to 3 days after major surgery and 20-50 after minor surgery.

Can patients with Hemophilia undergoing surgery be given continuous infusion regimes of

factor products? What are the necessary precautions to be taken while on continuous

infusion ?

Continuous infusions may provide improved coagulation factor cover, are associated with

improved bleeding outcomes and may use less coagulation factor than bolus regimes.

After an initial bolus injection of FVIII, an infusion is started to maintain coagulation according

to the following formula:

Infusion rate (IU/kg/h) = clearance (mL/kg/h) x steady state concentration (IU/mL)

Ideally, clearance can be calculated from pre-operative pharmacokinetic studies which can be

usedto guide initial infusion rates over several days.

Alternatively, an infusion rate of 3.0-5.0 IU/kg/h will produce a Factor level of

approximately 80 IU/dL. (See Table below for titrating the infusion regime)

Table 25.Guidelines for coagulation factor target levels in adults undergoing major surgery

using continuous infusion of recombinant factor VIII/IX

Day

Continuous Infusion

Target FVIII /IX steady state level

(%) Infusion rate dose (IU/kg/hour)

Pre-op(Bolus) 80-100 IU/kg load (round up to nearest vial size)

1-3 30-60 3.0-5.0 IU/kg/h

4-5 20-50 1.5.-4.0 IU/kg/h

5 and beyond Often change to bolus dosing

Page 407: Training Manual for Hemoglobinopathies and Hemophilia:

407

Continuous factor infusion must always be on an infusion pump. The following things should be

kept in mind:-

a. Always run a dedicated line

b. Factor should hang no longer than 12 hours, after 12 hours it should be

discarded and a new infusion set up.

c. Brand of the Factor product should always be checked for the use as continuous infusion. The

prerequisite is the factor should remain stable for the infusion hours after dissolution.

d. When one hour’s worth of factor left in the pump, a new infusion should be made ready

What are the preanaesthetic, intra-operative and post operative precautions to be kept in

mind for patients with bleeding disorders undergoing surgery?

The following factors need to be considered in Preoperative and Intraoperative and Postoperative

periods:-

Table 26. Preoperative anaesthetic assessment in patients with Hemophilia

History Examination Investigation

Nature of Bleeding disorder

Severity

Prior bleeding history

Prior treatment (factor therapy/Plasma

/transfusion etc/response to DDAVp)

Prior history of inhibitors Comorbidities

(especially HIV,HbsAg,Anti HCV) and

chronic liver disease or CD4 count

Availability of factor support and

haemophilia expert/haematologist

Dental hygiene and infections -treat

infection prior to orthopaedic surgery

Type of surgery:-

Elective-Always schedule on weekdays

Joint

deformities

Venous access

Airway

Blood Counts

Coagulation profile

including Fibrinogen

Specific Factor assay

if not already done

Inhibitor screen

within past 2 months

Ensure availability

of cross matched

blood products

Page 408: Training Manual for Hemoglobinopathies and Hemophilia:

408

with availability of full multidisciplinary

team)

Urgent-Ensure adequate factors

Table 27.Intraoperative Considerations during surgery in patients with Hemophilia

Peripheral lines are secured with utmost care.

Intramuscular injections and arterial punctures are avoided.

Smooth induction is given under deep plane of anesthesia.

Risk–benefits for neuraxial block and regional blocks need to be assessed individually and

in general are avoided.

To prevent muscle fasciculation (aggravate muscle and joint hematomas), use of

succinylcholine is avoided.

Utmost caution is done to do atraumatic tracheal intubation

Gentle suctioning of oropharynx should be done.

Extremities and pressure points should be padded.

Non-steroidal analgesics are not used as they cause gastrointestinal hemorrhage.

Hypothermia is avoided.

Hemodynamic conditions are maintained near normal,hypertension and tachycardia is

avoided

Small vessel hemostasis is achieved with minimal intraoperative blood loss

Tranexamic acid can be used at i.v dose of 10 mg kg 8th hourly). It should be avoided in

patients with haematuria due to obstructive uropathy.

Estimates of blood loss during surgery (Table 6) and data on pre- and post-operative

hemogloblin levels and the number of packed red blood cell units transfused should be

maintained to estimate surgical blood loss.

Surgical hemostasis should be assessed by an involved surgeon and/or anaesthetistand

Page 409: Training Manual for Hemoglobinopathies and Hemophilia:

409

Post-operative Considerations during surgery in patients with Hemophilia

Early mobilization, and physiotherapy should be instituted

Consider mechanical deep venous thrombosis prophylaxis using pneumatic

compression devices if available. Pharmacologic prophylaxis are best avoided

Adequate post operative analgesia should be given using tramadol, codeine and or

morphine. NSAIDs are avoided.

Drains, dressings and sutures should be removed at the time of peak factor

levels.(Within 1 hour of bolus dose of factor)

Patients with mild hemophilia A, as well as patients receiving intensive factor

replacement for the first time should be re-screened 4–12 weeks post-operatively

for development of inhibitors.

What are the precautions to be undertaken in female patients who are Hemophilia

Carriers while undergoing obstetric procedures / delivery?

Factor VIII and IX levels must be measured immediately before delivery and post-

partum with an aim to maintain above 0.5 IU ml(>50%) and accordingly supplemented

with factor VIII or Factor IX

Instrumental delivery is best avoided

The decision about caesarean section or vaginal delivery is as per obstetric indication

What are the precautions to be undertaken while undergoing dental procedures in patients

with Hemophilia ?

Close liaison between the dental surgeon and the hemophilia team is essential to provide

good comprehensive dental care.

Treatment can be safely carried out under local anesthesia using the full range of

techniques available to dental surgeons. Infiltration, intra- papillary, and intra-

records should be completed within 72 hours following surgery.

Page 410: Training Manual for Hemoglobinopathies and Hemophilia:

410

ligamentary injections, an inferior alveolar nerve block or lingual infiltrationare to be

done under factor cover of 20-40%.

Tranexamic acid or epsilon aminocaproic acid (EACA) is often used after dental

procedures to reduce the need for replacement therapy.

Following a tooth extraction, the patient should be advised to avoid hot food and drinks

until normal feeling has returned. Smoking should be avoided as this can cause problems

with healing.

Regular warm salt water mouthwashes (a teaspoon of salt in a glass of warm water)

should begin the day after treatment and continue for five to seven days or until the

mouth has healed.

What are the special precautions to be undertaken prior to undertaking orthopaedic

surgery in patients with Hemophilia?

Orthopedic surgeons should have had specific training in surgical management of persons

with hemophilia

If frequent bleeding is isolated to one target joint, minimally invasive procedures (e.g.

synovectomy) are preferred for young patients without significant joint damage.

Consider more aggressive and proactive strategies for patients in late teens to twenties as

they are more likely to have successful outcomes than older patients with significant joint

deterioration.

Consider patients with some preserved joint function and muscle mass as surgical

outcomes are more likely to be successful if joints are not already fixed with extensive

scar tissue and atrophied muscles.

Weigh benefits of multiple interventions and multiple site elective surgery using one

anaesthesia and favtor or bypassing agent coverage (cost saving) against the impact on

rehabilitation (easier vs. harder to rehabilitate and issues with assistive aids, such as

crutches) and stress to other arthritic joints.

What are the chances of inhibitor development during post operative period?

If the patient has had >50 exposure days to factor, the chances of developing inhibitor is

extremely low. Presence of inhibitor should be suspected if

Page 411: Training Manual for Hemoglobinopathies and Hemophilia:

411

There is unexplained bleeding despite adequate factor support

The trough levels of factor are below the expected range

A repeat Bethesda assay should be done if inhibitor development is suspected.

If inhibitor is confirmed to be present, switch to bypassing agents will be required.

Special situations

Patient with confirmed high titre inhibitor

In such a scenario, adequate FEIBA or rVIIa will be required . Since these are much

more expensive and in short supply compared to factor, adequate planning will be

required.

Factor assays will not be useful to monitor whether adequate haemstasis has been

achieved or not.

Tests of global haemostasis like TEG and Rotem might give some insight, however there

are no definite guidelines to modify dose based on the results.

Patients on newer agents

Newer agents like extended half life factors are useful in achieving surgical haemostasis

with the added advantage of less frequent doses.

This could vary from once a day for factor VIII products to once in 36-48 hours for factor

IX products, depending on the half life.

However standard factor assays might give falsely elevated or low level depending on the

reagents used. Therefore the assay should use reagents proven to give accurate factor

assay for the particular extended half life product.

Non factor agents like emicizumab and fitusiran are useful for prophylaxis of bleeds, not

for surgical hemostasis. However, if some one has already been on these agents and

requires emergency surgery, the dose of factor required will be much less.

If a patient with high titre inhibitor on emicizumab or fitusiran requires surgery, the dose

of bypassing agents used should be much less than standard recommendations.

In the above scenario, close monitoring should be done to look for thrombosis.

Suggested reading

Page 412: Training Manual for Hemoglobinopathies and Hemophilia:

412

1. Srivastava, A, Santagostino, E, Dougall, A, et al. WFH Guidelines for the Management of

Hemophilia, 3rd edition. Haemophilia. 2020: 26(Suppl

6): 1‐ 158. https://doi.org/10.1111/hae.14046

2. Srivastava A et alLow-dose intermittent factor replacement for post-operative

haemostasis in haemophilia.Haemophilia1998 ;4:799-801.

3. Mahlangu J etal Long-acting recombinant factor VIII Fc fusion protein (rFVIIIFc) for

perioperative haemostatic management in severe haemophilia

ThrombHaemost2016;116:1-8

4. Rickard KA. Guidelines for therapy and optimal dosages of coagulation factors for

treatment of bleeding and surgery in haemophilia. Haemophilia. 1995 Jan;1 Suppl

1(S1):8–13.

5. Mathews V, Viswabandya A, Baidya S, George B, Nair S, Chandy M, et al. Surgery for

Hemophilia in Developing Countries. Semin Thromb Hemost. 2005 ;31:538–43.

6. Solimeno LP, Escobar MA, Krassova S, Seremetis S. Major and Minor Classifications

for Surgery in People With Hemophilia: A Literature Review. Clin Appl Thromb 2018

;24:549–59.

Cautions to be exercised while using newer products/novel therapeutic

approaches for haemophilia

Newer therapeutics in hemophilia include the extended half-life (EHL) products and the recently

approved substitution therapy (Emicizumab) while a range of haemostatic rebalancing therapies

and gene therapy are in various stages of clinical development.

Newer Extended Half Life Products

Page 413: Training Manual for Hemoglobinopathies and Hemophilia:

413

The plasma half- life of conventionally used recombinant and plasma-derived therapies require

frequent administration in both therapeutic and prophylactic setting. The novel bio-engineered

extended half -life products with extended half-life enable less frequent dosing with similar

efficacy.

Table 28.Salient features of newer Factor VIII EHL products

Product

Source Half life Dosing interval Features FDA

Approval

NOVOEIGHT

CHO

cell line

10-12

hours

12-24 hours

P*-20-50 /kg IU

thrice weekly

Single use vial

Tyrosine sulphation

October

2013

ELOCTATE

HEK

cell line

19.7hours 24-48 hours

P-50 IU/kg q 96

hourly

Fc infusion protein

Infrequent inhibitors

June 2014

NUWIQ

HEK

cell line

17.1

hours

12-24 hours

P-40IU /kg IU q

48 hourly

Tyrosine sulphation September

2015

ADYNOVATE

CHO

cell line

13-15

hours

12-24 hours

P-40-50/kg IU

twice weekly

Pegylation

November

2015

KOVALTRY

BHK

cell line

14.3

hours

12-24 hours

P-20-50/kg IU

thrice weekly

Glycosylation and

tyrosine sulphation

March 2016

ESPEROCT

CHO

cell line

19 hours 12-24 Hours

50IU/Kg every 4

days

Pegylation February

2019

JIVI BHK

cell line

19 hours 12-24 hours P-

25IU/kg twice

weekly

Pegylation August

2018

AFSTYLA CHO

cell line

14.3

hours

12-24 hours

P-20-50/kg IU

thrice weekly

Single chain May 2016

*P-Prophylaxis

Table 29.Salient features of Factor IX EHL products.

Product Source T1/2 Dosing

frequency

Features FDA

Approval

RIXUBIS CHO cell line 25.4 hours P- 60- 80

IU/kg twice

weekly

Identical to

plasma derived

June

2103

Page 414: Training Manual for Hemoglobinopathies and Hemophilia:

414

ALPROLIX HEK cell line 86.5 hours P- 50 IU/kg

weekly

Fc fusion protein March

2014

IXINITY CHO cell line 24 hours P-50IU/Kg

weekly

Identical to

plasma derived

June

2015

IDELVION CHO cell line 87hours P- 50 IU/kg

Weekly

Albumin fusion March

2016

A. EHL Factor VIII (Extended Half Life factor VIII):

Eloctate

Indications:

o On demand treatment of bleeding episodes

o Perioperative management of bleeding

o Routine prophylaxis

Table 30.Dosing and frequency of on demand therapy Eloctate infusion

Table 31.Dosing and frequency of Eloctate for perioperative management

Type of bleed Factor VIII

levels

required (%

of normal)

Dose

(IU/Kg)

Frequency Duration of

therapy

Minor or moderate

bleeding (Joint/

muscle bleed,

mucous membrane

bleed)

40-60 20-30 Every 24-48 hours

(12-24 hourly in

patients <6 years)

Until bleeding is

resolved

Major bleeding

(Limb/ life

threatening bleed, IC

bleed,

Neurovascular

compromise)

80-100 40-50 Every 12-24 hourly

(8-12 hourly in

patients <6 years)

Until bleeding is

resolved

(approximately

7-10 days)

Page 415: Training Manual for Hemoglobinopathies and Hemophilia:

415

Type of surgery Factor VIII

levels required

(% of normal)

Dose

(IU/Kg)

Frequency Duration of

therapy

Minor

(Uncomplicated

tooth extraction)

50-80% 25-40 Every 24 hours (12-24

hourly in patients <6

years)

At least 1 day

until healing is

achieved

Major

(Neuro/abdominal

surgery/Joint

replacement)

80-120 (pre and

post-operative)

Preoperative:

40-60

Repeat: 40-

50

8-24 hourly

preoperatively.

Post operatively every

24 hourly

Until adequate

wound healing

Additional Information on Eloctate

o Cannot be used for von Willebrand disease

o Strengths: 250, 500, 750, 1000, 1500, 2000 and 3000 IU vials

o 1-5year age group has increased clearance of Eloctate, hence need higher or more frequent

dosing.

B. EHL Factor IX products

Alprolix

o Indications

a. On demand treatment of bleeding episodes

b. Perioperative management of bleeding

c. Routine prophylaxis

Table 32. Dosing and frequency of on demand therapy Alprolix infusion

Type of bleeding Circulating factor IX

level needed (% of

normal)

Dosing interval

Minor and Moderate:

Uncomplicated hemarthrosis,

superficial muscle

30-60 Repeat after 48 hours if there is

further evidence of bleeding

Page 416: Training Manual for Hemoglobinopathies and Hemophilia:

416

hematomas.

Major: Deep muscle

hematoma, neurovascular

compromise, CNS bleed,

Retroperitoneal bleed,

retropharyngeal bleed.

80-100 Consider a repeat dose after 6-10

hours and then every 24 hours for

the first 3 days. After 3 days

frequency may be adjusted every

48 hours or longer until bleeding

stops and healing is achieved

Additional Information on Alprolix

o Strength: 250, 500, 1000, 2000, 3000 and 4000 IU vials

o Not indicated for ITI

Cautions in laboratory monitoring of EHL products

The commonly used laboratory methods for measuring FVIII or FIX activity may not be

optimal for some modified rFVIII or

products.

The most consistent laboratory methods that can adequately measure modified rFVIII

products are based on chromogenic principles.

Any laboratory that measures factor activity in hemophilia patients should have knowledge

about (1) which replacement factor the patient is receiving and (2) whether their current

method(s) for determining factor activity provides an accurate (within 25% of target)

measurement of that particular replacement therapy.

For those laboratories that do not have the optimal measuring techniques, the treating

clinician must be informed about the potential biases (over- or underestimating factor

activity) and an alternative means of measuring accurate concentration provided.

C. Monoclonal antibody:Emicizumab (Monoclonal modified IgG4 with a bispecific antibody

structure) [ HEMILIBRA]

o Indications: Routine prophylaxis for bleeding episodes in patients with:

Hemophilia A with factor VIII inhibitors

Severe Hemophilia A without factor VIII inhibitors

Page 417: Training Manual for Hemoglobinopathies and Hemophilia:

417

Dosing: The total volume of Hemlibra to be injected subcutaneously is calculated as follows:

Total amount (mg) of emicizumab to be administered ÷ vial concentration (mg/mL) = total

volume of Hemlibra (mL) to be injected

Loading dose: 3mg/kg s/c once a week for first four weeks

Maintenance dose (from Week 5):

1.5mg/kg s/c once weekly OR,

3mg/kg s/c once in two weeks OR,

6mg/kg s/c once in four weeks.

Cautions:

Strengths available:

30mg/ml vial

60mg/0.4ml vial

105mg/0.7ml vial

150mg/ml vial

Treatment with bypassing agents (aPCC, rFVIIa){BPA} must be

discontinued 1 day before the start of therapy with hemlibra

Factor VIII PROPYLAXIS may be continued for the first 7 days of

therapy with hemlibra

The total volume of the dose must be injected subcutaneously only.(Not

intravenous or intramuscular)

Different hemlibra concentrations (30 mg/ml. 60 mg/ml) should not be

combined in the same syringe

A volume of >2 mL per injection should not be administered

Use of bypassing agents in patients who are on HEMILIBRA

Concomitant use of BPAs and hemlibra is a risk factor for thrombotic microangiopathy

(TMA) and thromboembolism because hemlibra increases the patient’s coagulation potential

BPAs must be discontinued 1day before the start of hemlibra therapy

If aPCC is indicated in patients on hemlibra, then the initial aPCC dose should not be

>50U/kg and laboratory monitoring is indicated (renal function, platelet count, evaluation

Page 418: Training Manual for Hemoglobinopathies and Hemophilia:

418

for thrombosis). The total dose of aPCC should not exceed >100 U/kg in the first 24 hours

of treatment

Recommendations on laboratory assays while on emicizumab

Although emicizumab mimics the function of activated factor VIII, it is biologically

different from activated factor VIII, having much lower affinity for factor IXa and X, not

requiring activation, and not deactivated by the protein C/S pathway.

These fundamental differences affect our interpretation of clotting assays

Laboratory monitoring of emicizumab is not required while on routine prophylactic dosing

APTT-based assays, including clot-based FVIII activity assays, should not be performed

while on emicizumab, as they will yield misleading results (ie. artifactually shortened aPTT

and elevated FVIII activity).

Chromogenic FVIII activity assays will only provide an assessment of emicizumab activity

if the assay includes all human reagents but these are not widely available. A chromogenic

FVIII assay that uses bovine reagents may be used to assay the FVIII activity of any

additional FVIII concentrate administered or endogenous FVIII.

The clot-based Bethesda assay cannot be utilized to assess FVIII inhibitor levels. However,

a bovine-reagent chromogenic-based inhibitor assay can report out FVIII inhibitor levels. If

samples are submitted to the CDC for central laboratory testing, they must be clearly

identified that the patient is on emicizumab.

Table 33.Coagulation tests affected and unaffected with Emicizumab

Results affected by Emicizumab

Results unaffected by Emicizumab

APTT - Bethesda assays (clotting-based) for

FVIII inhibitor titers

- One-stage, APTT-based, single-factor assays

- APTT-based activated protein C resistance

(APC-R)

- Activated clotting time (ACT)

-Bethesda assays (bovine chromogenic) for

FVIII inhibitor titers

- Thrombin time (TT)

- One-stage, prothrombin time (PT)-based,

single-factor assays

- Chromogenic-based single-factor assays other

than FVIII1

- Immuno-based assays (e.g. ELISA,

turbidimetric methods)

Page 419: Training Manual for Hemoglobinopathies and Hemophilia:

419

- Genetic tests of coagulation factors (e.g.

Factor V Leiden, Prothrombin 20210)

GlycoPEGylated recombinant factor IX (rFIX);

Indication : Treatment and prophylaxis of bleeding in pretreated patients with haemophilia B

(congenital factor IX deficiency).

Dosage:

Dosage

Prophylaxis Once weekly dosage – 40IU/kg

Adjustment of doses & administration intervals may be considered

based on achieved FIX levels & individual bleeding tendency

On-Demand Degree of

Haemorrhage

Recommended

dose IU/kg of

Refixia

Dosing recommendation

Early haemarthrosis

muscle bleeding or

oral bleeding. More

extensive

haemarthrosismuscle

bleeding or

haematoma

40 – 1 dose A single dose is

recommended

Severe or life

threatening

haemorrhages

80 – 1 dose Additional doses of

40IU/kg can be given

Surgery Minor surgery

including tooth

extraction

40 – 1 dose Additional doses can be

given if needed

Major surgeries 80 – 1 dose Pre-operative doses

40 – 3 doses 1st week - 2 repeat doses

(1-3 days interval)

2nd week onwards Once

weekly until bleeding

stops & healing achieved

Benefits :

Significantly reduces bleeding with a median ABR of 1 and median spontaneous ABR of 0in

adolescents and adults.

In children, median ABR of 0.66, and median spontaneous ABR of 0 was seen

Page 420: Training Manual for Hemoglobinopathies and Hemophilia:

420

Has the potential to prevent further bleeds in target joints by showing 100% resolution of target

joints at end of treatment

Less frequent injections:98.4% of bleeding episodes were controlled by 1 injection in

adolescents and adults and82% of bleedsin children.

Half-lifethat is five times longer than existing treatment meaning that Refixia® has the potential

to be incorporated into a once-weekly dosing schedule.

In patients undergoing surgery, a single preoperative dose provided effective haemostatic

coverage, and no patient required additional doses on the day of surgery.

Have a favourable safety profile with no inhibitorsor thromboembolic events or other adverse

effects

Significant improvement in the quality of life

Suggested reading

Weyand AC, Pipe SW. New therapies for hemophilia. Blood 2019 ;133:389 - 98.

Kitchen S, Tiefenbacher S, Gosselin R. Factor Activity Assays for Monitoring Extended

Half-Life FVIII and Factor IX Replacement Therapies. Semin Thromb Hemost. 2017

;43:331–7.

Page 421: Training Manual for Hemoglobinopathies and Hemophilia:

421

A Comprehensive model for conservative management in hemophilia

Hemophilia is known to cause bleeds in the joints and muscles, or bleeds in various systems of

the body-the brain, the renal system, the gastro-intestinal system. Bleeds may be spontaneous

bleed or due to injury. Every systemic bleed or bleed in the muscle or joint if not treated

appropriately and on time can lead to complications and compromise functioning.The

musculoskeletal [MSK] problems due to hemophilia are seen in the form of joint and muscle,

kinetic and kinematic dysfunction. The late effects on this system could be seen as-early joint

disintegration, joint deformities, loss in muscle mass, compromised muscle strength,

neurovascular complications, and pseudo tumors; ultimately a jeopardized function and quality

of life.

The PWH who do not receive adequate and consistent CFC have higher rate of mortality and

disability, most of the PWH suffer pain. In parts of the world where there is no or insufficient

factor supply the conservative management seems to be more affordable and feasible, and it is

this management that has to be have an evidence and scientific basis for management.Looking

at the international protocol of management of a bleed it is imperative to infuse factor in the

golden period after the bleed, that is ASAP, soon as the aura is experienced by the PWH, or

signs of bleed are noticed by the caregiver of the PWH in children who are very young.

Those on prophylaxis are seen to have lesser incidence of bleeds and an improved QoL versus

who are not on regular prophylaxis. Yet, there are incidences when the PWH on prophylaxis do

have bleeds due to one or the other reasons and in both [patients treated and untreated with

prophylaxis] the groups, issue remains managing an acute bleed. Especially for the group of

patients who are being managed on demand basis and have recurrent bleeds, immediate and

appropriate conservative management is essential.

Page 422: Training Manual for Hemoglobinopathies and Hemophilia:

422

I] Early infusion of factor concentrate: Thisis the most important aspect, however when not

possible to infuse, the managementmay start with step II.

Steps in management of an acute bleed when factor availability is not immediate or not

possible:

ii] Rest the joint and muscle

For joint bleeds, support the joint with a splint or rest the joint on a soft cushioned roll,

preferably in a position that relieves pain and stretch on the joint and then in the anti-deforming

positions, that is in the direction of extension range. This may be true depending on the site of

the bleed. For a flexed hip –knee position rest the knee on a roll under the knee so as to support

the hip in a pain free position.

In case of soft tissue bleeds ( muscle bleeds, compartment bleeds) which are seen on the body

surface, consider using a splint to the adjacent joint/s if deformed and splint the muscle

surface using a well-padded splint, covering a maximum area of bleed.

Infuse factor within the golden period

Before the bleed takes a large space

in the joint.

Before the joint temperature

Increases.

Soon as joint feels stiff or there is

difficulty in moving the joint.

Before the body part assumes a position

deviating from the normal alignment.

Benefits of early infusion

Reduces the chance of increase in

the bleed size and volume.

Enhances early absorption of the

bleed in the period following the

bleed.

Reduces pain.

Helps early mobility.

Helps early recovery and enhances

the healing process.

Figure 12: Resting in splint

Page 423: Training Manual for Hemoglobinopathies and Hemophilia:

423

Splints should be cost effective and

reusable.

Angle of the splint should be adjustable.

Depending on the site of bleed decision

to support a large surface area can be

made.

Consider splinting with a rigid material, but a soft and accommodative padding on the

surface that is in contact with the skin.

Splinting may be done using the three-point pressure principle, or may be simply

used as surface contact splints, with respect to the need.

Immobilization in splint helps to control unnecessary movements may prevent the clot

from dislodging.

Features of splinting

The joints have a tendency to recurrent bleeds, they can be used at the time of each

bleed.

With gradual resumption of the normal angle of joints, the splint angle should be

modifiable.

Rigid splints offer better rest; soft padding help to accommodate the change in joint

size in the splint, subsequent to the factor infusion.

The principle of three-point pressure can be more relevant to established anterior-

posterior deformities, where the intention is correction. Surface splints are options in

cases where there are muscle bleeds, or no joint static angulations observed.

Page 424: Training Manual for Hemoglobinopathies and Hemophilia:

424

III] Icing: Icing is the next step to be executed. Icing is a controversial approach, as evidence

proves thatsome patients are not able to tolerate icing due to aggravation of pain, some have

even reported about an increase in the size of the bleed. It is a myth that icing helps to clot

theblood, however it does not.

Procedure of icing

1.Do not use ice directly on the skin.

2.Crush and wrap ice in a thin cloth, cover the area to be cooled with a cloth, before

icing. Use ice cold water if icing cannot be tolerated.

3.Watch for skin color while icing.

4.Do not cool the part for more than 20 mins. Cold flex bandages could be preferred to,

if direct icing is not tolerated by the patient.

5. Use ice cold water alternated with normal room temperature water if cold water is not

tolerated. [Dose: 3 mins ice water/3 mins water at room temperature, repeat the procedure 5-

6 counts end with ice water].

6. Repeat the process two to three times a day if well tolerated.

Page 425: Training Manual for Hemoglobinopathies and Hemophilia:

425

Benefits of icing

1.It helps to cool the soft tissues and reduces the metabolic rate of tissues. Thus, reduces

the accumulation of the P ‘Factor at the site.

2.Icing can reduce pain in patients who tolerate it well, help to relax the muscle and

reduce the pain - spasm effect on the muscle

3.In advanced cases of arthropathy icing can aggravate pain, than reduce pain.

4.Watch for abrasions on the skin before soaking the wound with water due to condensation.

Page 426: Training Manual for Hemoglobinopathies and Hemophilia:

426

Figure 13. Use of ice cold water . If ice cubes are not tolerated cover skin with a thin cloth

or plastic while icing

IV] Compression: Compression can help better in immobilizing the part, application of

compression canalleviate pain, and reduce the swelling in the soft tissues. Do not continue with

tight, elastic, compression bandages for long after the bleed, especially over the muscle mass.

Constant and tight compression around the joint and soft tissues adds to aggravate muscle

wasting, habitual joint positioning and dependence.

How and when to use compression Benefits and ill effects

1.Continued compression under pressure

can cause muscle wasting and makes

muscle weak in the long run, due to

disuse atrophy.

2.Padded and protective cuffs should be a

better option to tight elastic wraps or tubi-

grips. As the padded cuffs allow optimal

contraction of the muscle fibers in

functional ranges, do not hold the joint and

muscle in static positions.

3.There is equal distribution of pressures

around the joint and on the muscle, in the

padded cuffs.

4. Padded cuffs provide protection and

proprioceptive input while the joint is

used.

1.Helps to support the joint during an

acute bleed.

2.Compression applied to the joint

helps to control the edema; in the soft

tissue helps reduce edema.

3.Compression helps to alleviate pain in

the acute stage.

4.It is best to apply compression with a

supporting splint as described above [ in

splinting], so as to avoid any deleterious

effects of external pressures, added to the

internal pressure caused by the bleed.

5.Do not continue to use compressionbandages

continuously after the bleed and pain subside.

Page 427: Training Manual for Hemoglobinopathies and Hemophilia:

427

IV] Elevation and Exercises: Elevating a joint to reduce the size of bleed may not be a good

option, especially if the bleed in the joint is in the intra-articular i.e. in the within the articular

capsule and not in the extra-articular space. For the muscle bleed elevation may be a good option

provided it is in the peripheral musculature, in deep muscle bleeds elevation may not be a

possible e.g.in case of the psoas bleed. In joint bleeds if there is effusion in the soft tissues

around the joint elevation can help reducing the swelling. Take care to keep the elevated part

above the level of heart when in supine lying.The `E’ also stands for exercises following a bleed;

Exercises should be started after appropriate and adequate amount of factor infusion, and

depending on the patients response to factor infusion. Exercises should be done in the expert

hands and under expert supervision of an MSK expert who may be a trained .

COMPRESSION

Figure 14: Compression.

Exercises have to be done after appropriate replacement of the CFC, after the pain subsides, and

after the joint movement can be initiated with support and assistance to the limb.

Initially focus on static isometric exercises in order to allow muscle fiber contractions with the

objective to prevent disuse atrophy.

Focus on ROM exercises so as to allow joint ranges to return to pre-episodic levels.

Exercises have to continue progressively to improve the muscle mass and strength.

Train the care givers to do simple exercises with hands on training so as to continue with home

programs along with home based prophylaxis.

Timely correction of compensated postures, exercise the agonist and antagonists to establish

symmetric balance of musculature.

Core muscle training, proprioceptive training to lower extremity joints for acquiring a near

normal gait pattern, fall prevention is an essential part of rehabilitation.

Weight bearing, static isometric contractions exercises to prevent bone demineralization are

equally important.

Page 428: Training Manual for Hemoglobinopathies and Hemophilia:

428

Occupational Therapist or a Physical Therapist in management of hemophilia care. Care givers

of the PWH may be trained for simple exercises that can be continued at home.

.

V] The other aspects of conservative management are complexities of haemophilia and need to

be dealt with, but individually are :

Figure 15: Muscle wasting

secondary to recurrent shoulder

bleed

Figure 16: Postural deviation due to

recurrent bleeds

Page 429: Training Manual for Hemoglobinopathies and Hemophilia:

429

Management of synovitis.

Management of patients with inhibitors.

Management of pseudo tumors and established

hematomas.

Management of psoas bleeds.

Management of soft tissue bleeds with residual

complications. Pain management.

Assessment tools for QoL.

Psycho-social aspects of the PWH related to the physical

limitations. Therapeutic intervention at different stages of the

arthropathy.

Conservative management of contractures and

deformities. Post joint replacement therapeutic

intervention.

Applicability of physical agent modalities in

hemophilia.

Compensatory devices for upper and lower extremity

adaptations. Clinical assessments and approaches to joint

mobilization.

Suggested Reading

Srivastava A, Brewer AK, Mauser-Bunschoten EP, Key NS, Kitchen S, Llinas A et al; Treatment

Guidelines Working Group on Behalf of The World Federation OfHemophilia. Guidelines for

the management of hemophilia.Haemophilia. 2013 ;19:e1-47

Page 430: Training Manual for Hemoglobinopathies and Hemophilia:

430

Psychosocial care and support: Training curriculum for Psychologists/Social

Workers/Nurses working with patients with bleeding disorders

Psychosocial impact in hemophilia is a lifelong condition affecting the PWH as well as the

immediate family members, friends and relatives. It affects them socially, physically and most

important emotionally/psychologically. Hemophilia is a not very common condition and as a

result, a large number of people are unaware about it. Specifically, Hemophilia population's need

of psychosocial support is underestimated due to myths, stigma and lack of knowledge.

However, psychosocial care and support in hemophilia are interventions and methods that

enhance the ability of children, families with Hemophilia, and communities to cope, in their own

context, and to achieve personal and social well-being; enabling PWH to experience love,

protection, and support that allows them to have a sense of self-worth and belonging with better

quality of life.Such interventions and methods are essential in order for children to learn, develop

life skills, participate actively, and have faith for the future.

Principles of Psychosocial Support (PSS) Program

There are different effective and sustainable ways to offer psychosocial support to PWH and his

family. However, the main principles of PSS program are not only important but also extremely

helpful for PWH in the following way:

* A Psychosocial Program as an integral part of the Comprehensive Care' program of the

hemophilia care centre plays an important role in taking in to account the developmental stages

of the child.

* The program acknowledges the critical role that potential care-givers play in the well-being of a

PWH and his family.

* The program offers continuity upon which the PWH can rely on. Follow up, support and

supervision are all of critical importance.

Page 431: Training Manual for Hemoglobinopathies and Hemophilia:

431

* Program focuses on empowering the PWH and his family, improving their self esteem,resilience

and enhancing their life skills.

* Under the program, assistance is also provided to enable the PWH and the family members to

ask for help when they need it and know whom to approach. This also includes putting in place a

referral system with qualified professionals.

* It also provides opportunities to promote the formation of support groups and encourage

interaction amongst them.

* The program educates and sensitizes the community in order to effectively seek participation for

help and support.

* The program identifies and trains a group of volunteers/lay counsellors from the affected

population and provide them back-up support in the form of a mental health expert.

Psychosocial tools and techniques

There can be various tools and techniques, such as psycho-education, counselling, groups for

children support, youth and women, relaxation techniques, behaviour management, stress

management, building coping skills and resilience life Skill Education, goal setting, self-esteem,

confidence building, Loss and grief etc. Each of these tools will be dealt in detail depending on

the requirement.

The Psychosocial Support Training Curriculum

This curriculum is designed to improve quality of care for people with bleeding disorders and to

increase their quality of life by making psychosocial care an integral part of a multi-disciplinary

approach to healthcare.

Aims

Raise awareness of psychosocial issues that affect people with bleeding disorders and

their families

Advocate nationally for comprehensive care

Ensure psychosocial participation in multidisciplinary hemophilia care teams

Provide support, information, and contacts for psychosocial workers

Objectives of Psychosocial Curriculum

The objectives of the Psychosocial Training Curriculum are:

To offer important psychosocial educational materials (sessions) for teaching and

training purposes

Page 432: Training Manual for Hemoglobinopathies and Hemophilia:

432

To design “user friendly” information curriculum that can be adapted culturally

To provide assistance to the access and understanding of the material

The Psychosocial Training Curriculum Modules

The modules are teaching tools for educating the psychosocial care professionals on the topics

provided in this Psychosocial Curriculum. The modules are identified and selected as important

topics for patients and families.

The Individual section of each module contains learning objectives, a short description, and

suggested supporting resources if any; to provide to participants.

Learning Objectives

Each module in the Psychosocial Curriculum has specific learning objectives that we hope to be

accomplished by the end of the module. It is important for the trainer to teach to the learning

objectives allowing for cultural modifications and any material adaptations to the targeted

audiences.

Interactive Element

It is helpful and valuable to the sessions by adding an interactive component that can be used in

the session presentation. The interactive elements are outlined in the sessions and are available

for the presenter to access. Interactive learning is very effective as a tool for teaching. One can

adapt the interactive piece as needed to gain the best learning experience for the audience.

Following are the important topics that forms the training curriculum under the section of

Psychosocial Training Curriculum:

Team Approach to Hemophilia Care

Coping with Chronic Pain

Assessing and enhancing coping strategies (resilience)

Adherence

Education and Employment

Hemophilia: Psychosocial impact on Carriers

Genetic Counselling -Basic level

Sexuality through the Life Stages

Coping with complications – HCV, HIV, Inhibitors, & Joint Disease

Disclosure

Page 433: Training Manual for Hemoglobinopathies and Hemophilia:

433

Women and Teenage Girls with Bleeding Disorders

Quality of Life Assessment Tools

Each module has been described as follows:

Module 1: Team Approach to Hemophilia Care

Estimated Duration

The presentation length is 30 minutes, allow additional time for questions

Description

The module discusses the importance of Comprehensive Care in the treatment of Hemophilia

and bleeding disorders with an emphasis on the composition of the care team. It describes the

role of Social Workers/psychologists/Nurses in Hemophilia Care and the various activities of

HTC and the foundation in general with an example to how to make a Psychosocial Action Plan

for their respective work places.

Learning Objectives

At the end of this session, participants will be able to:

Understand the comprehensive care model in the treatment of hemophilia

Discuss psychosocial care in hemophilia

Understand the role of social workers/psychologists/nurses in hemophilia care

Recall the goals of the social worker/psychologist/Nurses

Examine an example of a psychosocial action plan

Module 2: Coping with Chronic Pain

Estimated Duration

The presentation length is 30 minutes, allow additional time for questions.

Description

This module provides literature-based information which explains the physical and psychological

components of pain and offers practical suggestions on how to cope with chronic pain.

Topics covered include: the role of exercise, aids to relaxation, recognizing stress, depression,

physical and psychological dependency, complementary therapies, and tips for the family and

peer support.

Learning Objectives

At the end of this session, participants will be able to:

Describe the physical and psychological components

of pain

Page 434: Training Manual for Hemoglobinopathies and Hemophilia:

434

Understand how to cope physically and psychologically with chronic pain

Module 3: Quality of Life Assessment Tools

Estimated Duration

Presentation length for the introductory slide deck is 30 minutes, allow additional time for

questions.

Presentation length for the advanced slide deck is 45 minutes, allow additional time for

questions.

Description

There are two versions one is “introductory” (30 minute presentation) and the other is

“advanced” (45 minute presentation). Considerable flexibility is built into both.

Both the introductory and advanced sections introduces the audience to complications, from

bleeding and/or treatment, patients with hemophilia (PWH) experience. These complications

include: joint disease, inhibitors, and past exposure to viral infections prior the availability of

purified factor products. The potential impact of each complication in multiple physical and

mental health Quality of Life (QoL) domains is discussed. A higher level of detail is provided in

the advanced deck, especially related to social and psychosocial issues.

Learning Objectives

At the end of this session, participants will be able to:

Enhance participant understanding of the importance of quality of life (QoL) assessment

tools

Demonstrate the difference between types of outcome measures, with a specific emphasis

on patient-reported outcome measures

Enhance participant understanding of the dimensions and determinants of health-related

(HR) QoL assessment

Raise participant awareness of types of HRQoL questionnaires, especially hemophilia-

related QoL instruments

Demonstrate criteria for selecting an instrument

Highlight advantages of using multipronged assessment tools

Identify key QoL influences in the bleeding disorders community

Page 435: Training Manual for Hemoglobinopathies and Hemophilia:

435

Module 4: Assessing and Enhancing Coping Strategies (Resilience)

Estimated Duration

The presentation length is 30 minutes, allow additional time for questions

Description

This module discusses coping strategies both positive and maladaptive. It will provide some

examples and tips to for individuals, families and the community as a whole on coping skills.

The concept of resilience is discussed as a path to empowerment and a positive coping skill.

Learning Objectives

At the end of this session, participants will be able to:

Identify and define types of coping strategies

Describe stressors

Recognize maladaptive coping

Irrational belief systems

Describe positive coping skills

Adaptive coping skills

Provide tips for patients and families on coping within the larger community and

extended family

Recall psychosocial counselling options

Individual versus group counselling

Understand resilience as a path to empowerment

Recall assessment tools

Community psychosocial support

Module 5: Adherence

Estimated Duration

The presentation length is 45 minutes, allow additional time for questions

Description

This module provides literature-based information related to adherence in persons with

hemophilia (PWH). Potential barriers, primary motivators, and strategies to enhance adherence

are key components of this module. To enhance audience participation, assess baseline

knowledge, and to gain insight into adherence-related issues in the country, there are questions to

the participants.

Page 436: Training Manual for Hemoglobinopathies and Hemophilia:

436

Learning Objectives

At the end of this session, participants will be able to:

Understand issues related to adherence

Assess and address potential barriers to adherence for both providers and

patients/families

Understand the potential impact of cultural influences on adherence

Identify tools and psychosocial strategies to assess and enhance adherence

Module 6:Education and Employment

Estimated Duration

The presentation length is 60 minutes it is likely that a longer time will be necessary depending

on the Interactive elements, allow additional time for questions

Description

Topics covered in the Education section of the Module include: challenges, disclosure, educating

schools, Information Sheets for schools, the physical education class, after a bleed, the student

with arthritis, dealing with absences, overprotectiveness and bullying. Topics covered in the

Career Planning and Employment section of the Module include: career planning, job seeking,

developing a Curriculum Vitae, interview skills, networking, volunteering and work experience.

Within the education section of this Module, a Group Work interactive element is included in

order for participants to identify the schooling and education challenges, issues and problems for

two age groups – those 4 – 12 years and those aged 12 – 18 years within their local context.

Within the employment section of this Module, there is an option to interview a person with a

bleeding disorder from the local community to speak about their experience with employment.

Learning Objectives

At the end of this session, participants will be able to:

Understand the importance of education and employment for those with bleeding

disorders

Describe the challenges surrounding education for those with bleeding disorders

Identify strategies to promote participation in education for those with bleeding disorders

Describe the issues and challenges for those with bleeding disorders with regards to

employment

Page 437: Training Manual for Hemoglobinopathies and Hemophilia:

437

Identify strategies to assist those with bleeding disorders in career planning, job seeking

and employment

Module 7: Psychosocial Impact on Carriers

Estimated Duration

The presentation length is 60 to 90 minutes, allow additional time for questions

Description

The module on psychosocial impact on carriers is a very interactive module which defines the

types of Carriers and what it means. Then it discusses the Issues regarding testing and when to

test with example from developed and developing countries. It has a component of group work

which helps the individual to identify the centre specific issues and discussion thereof. The

module also discusses the stigma, emotional and concerns on being a carrier. It also touches

upon the Genetic Counselling and availability of reproductive choices with the factors affecting

reproductive choices & informed decision and cope mechanism to deal with the situation.

Learning Objectives

At the end of this session, participants will be able to understand:

Types of carriers and what this means

Issues regarding testing and when to test

Psychosocial issues and concerns about being a carrier

Genetic counselling and availability of reproductive choices

Factors affecting reproductive choices and informed decision

Coping with the situation

Module 8: Genetic Counselling

Estimated Duration

The presentation length is 45 minutes, allow additional time for questions

Description

The genetic counselling module is in two parts Basic and Advanced. The basic module defines

the genetic counselling and the role of genetic counsellor vis a vis psychosocial worker. It gives

the information about the inheritance of haemophilia and basic information relevant to

Page 438: Training Manual for Hemoglobinopathies and Hemophilia:

438

counselling a patient/family about hemophilia. It also makes the participants recognize the

cohesiveness between genetics and other health care providers within the HTC.

The module sheds light on how genetic counselling should take into account a patient’s

experience and perception, as well as the influence of social, cultural, and religious contexts,

whatever the level of facilities and services.

The advance module talks about how to draw the family tree and its interpretations as well as

availability of reproductive choices to the client.

Learning Objectives

At the end of this session, participants will be able to:

Understand the role of a genetic counsellor in the HTC

Recognize cohesiveness between genetic counselors and other health care providers

within the HTC

Identify key topics relevant to counselling a patient/family about hemophilia including

inheritance, factors that influence decision making, and family dynamics

Module 9A: Life Stages

Estimated Duration

The presentation length is 30 minutes, allow additional time for questions.

Description

This module provides literature-based information which explain the key issues, objectives and

strategies of each developmental stage from birth to 65 years. Topics covered include: -

Parents coping and normalizing, building trust, acceptance of hemophilia and emotional

reactions;

Child’s feelings, confidence and coping, understanding treatment and acceptance;

Teenager - learning to self-infuse, independence and responsibility, compliance issues, education

and career planning;

Young adult – transition, working, dating and relationships, genetic counselling;

Middle adulthood – career and family development, peer support, dealing with limitations;

Maturity – slowing down, age of wisdom.

There are notes for the trainers under some slides.

Page 439: Training Manual for Hemoglobinopathies and Hemophilia:

439

Learning Objectives

At the end of this session, participants will be able to understand the key issues and strategies for

each stage of psychosocial development

Module 9B: Sexuality

Estimated Duration

The presentation length is 20 minutes; allow additional time for questions.

Description

This module provides literature-based information which explain the key issues and strategies

regarding sexuality throughout the life stages. The trainer’s local knowledge of cultural

sensitivity, can decide which topics to include or exclude.

Topics covered include: early years, gratifying body experiences, attitudes to masturbation,

puberty and adolescence, body image, communication, stress and the libido, sexual intercourse

and bleeds, effects on sexual desire, sexual expression and relationships.

Learning Objectives

At the end of this session, participants will be able to understand key issues and strategies

regarding sexuality through the life stages

Module 10: Coping with Complications – HCV, HIV, Inhibitors and Joint Disease

Estimated Duration

The presentation length is 30 minutes (Introductory), allow additional time for questions. The

presentation length is 45 minutes (Advanced), allow additional time for questions.

Description

There are two versions of this module, one is “introductory” and the other is “advanced”.

Considerable flexibility is built into both. The presenter can opt to exclude any complication(s)

due to time constraints or to lack of relevance for the particular audience, without impacting the

flow of the presentation.

Both the introductory and advanced sections introduce the audience to complications, from

bleeding and/or treatment, patients with hemophilia (PWH) experience. These complications

include: joint disease, inhibitors, and past exposure to viral infections prior the availability of

purified factor products. The potential impact of each complication in multiple physical and

mental health Quality of Life (QoL) domains is discussed. A higher level of detail is provided in

the advanced deck, especially related to social and psychosocial issues.

Page 440: Training Manual for Hemoglobinopathies and Hemophilia:

440

Learning Objectives

At the end of this session, participants will be able to:

Identify/list hemophilia-related complications

Describe at least three ways these complications may impact the psychosocial

functioning of patients/families/caregivers

Appreciate how complex the psychosocial impact of these complications can be for

patients/families

Identify and describe psychosocial intervention strategies

Module 11: Disclosure

Estimated duration

The presentation length is 45 minutes, allow additional time for questions.

Description

This is a two-part session on decision making around disclosing a diagnosis of hemophilia,

including who, when, where, and why to tell.

Learning Objectives

At the end of this session, participants will be able to:

Appreciate the complexity of issues related to disclosure

Understand disclosure within the context of the participants’ culture

Assess and address potential risks and benefits to disclosure

Identify three psychosocial strategies to facilitate disclosure decision making

Module 12: Women and Teenage Girls with Bleeding Disorders

Estimated Duration

The presentation length is 20 minutes, allow additional time for questions.

Description

This module provides literature-based information which explain the key issues for women and

teenage girls with bleeding disorders. The trainer’s local knowledge of cultural sensitivity, can

decide which topics to include or exclude.

Topics covered include: quality of life, genetic counselling, carrier testing, emotional reactions

of carriers, coping with emotions, tips for teenage girls, living positively with bleeding disorders.

Page 441: Training Manual for Hemoglobinopathies and Hemophilia:

441

Learning Objectives

At the end of this session, participants will be able to:

Understand key issues for women and teenage girls with bleeding disorders

The above modules can be used, adapted depending upon the audience, their levels and the

training duration.

Evaluation process

Conducting an evaluation process for instance, “ Evaluation Form” is a key element to the

success and outcomes of the program. Without collecting feedback or an evaluation, it is very

difficult to have information that is useful to the facilitating/training team to see if they have

accomplished their goals and for future improvement.

Page 442: Training Manual for Hemoglobinopathies and Hemophilia:

442

Blood Collection Transportation Vans (BCTV)

IICCHHHH

((IInntteeggrraatteedd CCeenntteerrss ffoorr

HHeemmoogglloobbiinnooppaatthhiieess &&

HHeemmoopphhiilliiaa))

IT solution to connect, digitize and streamline the

blood banks

Training for Hemoglobinopathies

and Hemophilia