Top Banner
1 PROPOSAL FOR THE INCLUSION OF BENZNIDAZOL PEDIATRIC DOSAGE FORM AS TREATMENT FOR CHAGAS DISEASE IN CHILDREN YOUNGER THAN 2 YEARS OLD IN THE WORLD HEALTH ORGANIZATION MODEL LIST OF ESSENTIAL MEDICINES FOR CHILDREN Drugs for Neglected Diseases initiative (DNDi) Rio de Janeiro, Brazil Persons to Contact: Isabela Ribeiro Head of Chagas disease programme Drug for Neglected Diseases initiative Latin America Santa Heloísa Street, 05 CEP: 22460-080 - Jardim Botânico Rio de Janeiro - Brazil Phone: +55 21 2215-2941 Email: [email protected] Gabriela Costa Chaves Drug for Neglected Diseases initiative Latin America Santa Heloísa Street, 05 CEP: 22460-080 - Jardim Botânico Rio de Janeiro - Brazil Phone: +55 21 2215-2941 Email: [email protected]
66

proposal for the inclusion of benznidazol pediatric dosage

Feb 12, 2022

Download

Documents

dariahiddleston
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: proposal for the inclusion of benznidazol pediatric dosage

1

PROPOSAL FOR THE INCLUSION OF BENZNIDAZOL PEDIATRIC DOSAGE FORM AS TREATMENT

FOR CHAGAS DISEASE IN CHILDREN YOUNGER THAN 2 YEARS OLD IN THE WORLD HEALTH

ORGANIZATION MODEL LIST OF ESSENTIAL MEDICINES FOR CHILDREN

Drugs for Neglected Diseases initiative (DNDi)

Rio de Janeiro, Brazil

Persons to Contact:

Isabela Ribeiro Head of Chagas disease programme Drug for Neglected Diseases initiative Latin America Santa Heloísa Street, 05 CEP: 22460-080 - Jardim Botânico Rio de Janeiro - Brazil Phone: +55 21 2215-2941 Email: [email protected]

Gabriela Costa Chaves Drug for Neglected Diseases initiative Latin America Santa Heloísa Street, 05 CEP: 22460-080 - Jardim Botânico Rio de Janeiro - Brazil Phone: +55 21 2215-2941 Email: [email protected]

Page 2: proposal for the inclusion of benznidazol pediatric dosage

2

Sumário

Glossary:..................................................................................................................................... 3

Executive Summary .................................................................................................................... 4

1. Summary statement of the proposal for inclusion, change or deletion ............................... 6

2. Name of the focal point in WHO for this application ........................................................... 6

3. Name of the organization(s) consulted and/or supporting the application .......................... 6

4. International Nonproprietary Name (INN, generic name) of the medicine .......................... 6

5. Formulation proposed for inclusion; including adult and pediatric (if appropriate) ............. 6

6. International availability - sources, if possible manufacturers and trade names .................. 7

7. Whether listing is requested as an individual medicine or as an example of a therapeutic group .......................................................................................................................................... 8

8. Information supporting the public health relevance (epidemiological information on disease burden, assessment of current use, target population) .............................................................. 8

8.1 Epidemiological information on disease burden ................................................................ 8

8.2 Assessment of current use .............................................................................................. 14

8.2.1 Target population..................................................................................................... 19

9. Treatment details (dosage regimen, duration; reference to existing WHO and other clinical guidelines; need for special diagnostics, treatment or monitoring facilities and skills) .............. 26

9.1 Dosage regimen, duration ............................................................................................... 26

9.2 Reference to existing WHO and other clinical guidelines ................................................. 26

9.3 Need for special diagnostic or treatment facilities and skills ........................................... 27

10. Summary of comparative effectiveness in a variety of clinical settings: .......................... 27

10.1 Identification of clinical evidence and summary of available data ................................. 28

10.1.4 Conclusions on efficacy .............................................................................................. 38

11. Summary of comparative evidence on safety*: .................................................................. 40

11.1 Safety Overview ........................................................................................................... 40

11.1.1 Introduction and Overview ........................................................................................ 40

11.2 Safety and Tolerability of Benznidazole ......................................................................... 41

11.3 Adverse effects during treatment with Benznidazole .................................................... 41

11.4 Adverse events in adults ............................................................................................... 44

11.5 Adverse events in children ............................................................................................ 45

11.5.1 Children with congenital infections ........................................................................ 45

11.5.2 Children with acute infections ................................................................................ 46

11.5.3 Children with indeterminate chronic infections ...................................................... 46

11.6 Conclusions on Safety and Tolerability .......................................................................... 54

12. Summary of available data on comparative cost** and cost-effectiveness within the pharmacological class or therapeutic group: ............................................................................ 55

13. Summary of regulatory status of the medicine (in country of origin, and preferably in other countries as well)...................................................................................................................... 56

14. Availability of pharmacopoeial standards (British Pharmacopoeia, International Pharmacopoeia, United States Pharmacopoeia) ....................................................................... 57

15. Proposed (new/adapted) text for the WHO Model Formulary ............................................ 57

List of Appendix ........................................................................................................................ 59

References ............................................................................................................................... 60

Page 3: proposal for the inclusion of benznidazol pediatric dosage

3

Glossary:

ANVISA: Brazilian Drug Regulatory Authority (Agência Nacional de Vigilância Sanitária, in

Portuguese)

API: Active Pharmaceutical Ingredient

BPCRS: British Pharmacopoeia Chemical Reference Substances

Bz: Benznidazole

CDC: Centers for Disease Control and Prevention

CFT: Complement Fixation Test

DNA: Deoxyribonucleic acid

DNDi: Drugs for Neglected Diseases initiative

EIA: Enzyme Immuno Assay

ELISA: Enzyme-Linked ImmunoSorbent Assay

Hc: Hemoculture

HIV/AIDS: Human Immunodeficiency Virus/Acquired Immunodeficiency Syndrome

ICRS: International Chemical Reference Substances

igG: Imunoglobulin G

IgM: Imunoglobulin M

IHA: Indirect Hemagglutination Assay

IIF: Indirect Immunofluorescence

INN: International Nonproprietary Name

IRD: Institut de Recherche pour le Développement

LAFEPE: Brazilian Pharmaceutical Laboratory of the State of Pernambuco (Laboratório

Farmacêutico do Estado de Pernambuco, in Portuguese)

MH: Microhematocrit

MSF: Doctors Without Borders (Médecins Sans Frontières, in French)

Nx: Nifurtimox

PAHO: Pan American Health Organization

PCR: Polymerase Chain Reaction

RENAME: Brazilian Essential Medicines List (Relação Nacional de Medicamentos Essenciais, in

Portuguese)

SAE: Serious Adverse Event

T. cruzi: Trypanosoma cruzi

TDR: Tropical Disease Research

TPP: Target Product Profile

UNICEF: United Nations International Children's Emergency Fund

USA: United States of America

USP: United States Pharmacopeia

WHA: World Health Assembly

WHO: World Health Organization

WS: Work Standard

Page 4: proposal for the inclusion of benznidazol pediatric dosage

4

Executive Summary

Benznidazol (Bz) pediatric dosage form (tablet 12.5mg) is proposed for inclusion in the World

Health Organization (WHO) Model List of Essential Medicines for Children, section 6.5.5.2, as

new dosage form indicated for children below 2 years old with Chagas disease.

A recent report published by WHO on Neglected Tropical Diseases in 20101 estimates that 10

million people worldwide are infected by T. cruzi, mostly in the endemic areas of 21 Latin

American countries, but also including non-endemic countries, as a consequence of population

mobility. An estimate published by Pan American Health Organization (PAHO) in 2006, indicates

a prevalence of approximately 8 million infected individuals and an incidence of 55,185 new

cases each year, among which 14,385 are congenital infections. Despite advances over recent

years in the control of vector and transfusions related Chagas disease, there is agreement that

congenital transmission is likely to remain of importance in the decades to come.

The treatment of Chagas disease is recommended for all cases of acute, congenital and reactive

infections among all children, and for patients up to 18 years old in the indeterminate chronic

phase (PAHO, 1998; WHO, 2002, 2011). Most infections occur during childhood, involving

children, including new-borns who are diagnosed at birth.

Notwithstanding existing recommendations for treatment of children, until recently the only

registered dosage form was an adult tablet of 100 mg. For years, the inadequacies of the current

form of administration and need for alternative formulations have been a consensus of among experts.

This submission for inclusion of the new dosage form of benznidazole is thus presented in the

context of an unmet medical need where it is held that the benefit of using this new 12.5 mg

benznidazole strength minimizes the risk of continued use of extemporaneous formulations of

the present 100 mg tablet for the pediatric population, and notably the very young.

Benznidazole is highly efficacious for the treatment of congenital infections. Seroconversion

rates vary from 87% (Schijman et al., 2003) at 36 months (100% in the 0 to 3 months old group)

to 100% (Russomando et al., 1998) at 24 months in children up to two years old. For the

treatment of acute infections, these rates vary from 76% (Cançado et al., 2002) at 13 years in

children under 10 years old to 100% (Ferreira, 1988) at 15 years in children between 2 and 18

years old.

Safety data summarized in this submission indicates a better tolerability for benznidazole

among children than adults, with treatment discontinuation due to adverse events being rare

among new-borns, and no greater than 10% in children in the indeterminate phase, while these

rates may rise to 40% among adults, usually hovering at around 20%.

The assessment of risk-benefit ratio of the use of the new strength of 12.5 mg versus the

continued use of the 100 mg tablet in the pediatric population points to the benefit of

prescribing a known concentration of the product, with easier management in the conditions

use, by parents and health professionals. The new strength of benznidazole in the form of

rapidly disintegrating tablet, would allow an easy and reliable administration, especially in long-

term home care.

Page 5: proposal for the inclusion of benznidazol pediatric dosage

5

pediatric

Page 6: proposal for the inclusion of benznidazol pediatric dosage

6

6

1. Summary statement of the proposal for inclusion, change or deletion

Benznidazol (Bz) pediatric dosage form (tablet 12.5mg) is proposed for inclusion in the World

Health Organization (WHO) Model List of Essential Medicines for Children, section 6.5.5.2, as

new dosage form indicated for children by 2 years old with Chagas disease.

2. Name of the focal point in WHO for this application

Pedro Albajar Viñas - WHO / HTM / NTD / IDM / Chagas disease

World Health Organization/Organisation mondiale de la Santé

HIV/AIDS, Tuberculosis, Malaria and Neglected Diseases

Control of Neglected Tropical Diseases

Innovative & Intensified Disease Management

Avenue Appia 20 - 1211 Genève 27 - Suisse

Tel.: +41 (0)22 7911261; fax: +41 (0)22 7914777

http://www.who.int/neglected_diseases

[email protected]

3. Name of the organization(s) consulted and/or supporting the application

Brazilian Ministry of Health – Appendix 1

Honduras Ministry of Health – Appendix 2

Argentinean Ministry of Health – Appendix 3

Médecins Sans Frontières – Appendix 4

Bolivian Ministry of Health – Appendix 9

4. International Nonproprietary Name (INN, generic name) of the medicine

Benznidazolum

Benznidazole

N-benzyl-2-nitroimidazole-1-acetamide

C12H12N4O3

Source: International Nonproprietary Names for Pharmaceutical Substances, 19742

5. Formulation proposed for inclusion; including adult and pediatric (if appropriate)

The formulation of pediatric benznidazole (12.5mg) has the same qualitative composition

and manufacturing process of the adult (100mg) registered by LAFEPE manufacturer(1), with

an adjustment on the proportion of diluents (starch and lactose). This adjustment was

needed due to the quantity of benznidazole contained in each dosage (8 times difference)

and the appropriate size of tablets, as shown in table 1.

Page 7: proposal for the inclusion of benznidazol pediatric dosage

7

7

Table 1: Formulation of adult (100mg) and pediatric (12.5mg) benznidazole

FORMULATION PEDIATRIC (%)

12.5 mg Bz

ADULT (%)

100 mg Bz

Benznidazole 10.00 40.00

Starch 51.20 31.20

Lactose 200 36.80 26.80

Magnesium Stearate 0.40 0.40

Talcum 1.60 1.60

Average weight of tablets (mg) 125.00 250.00

Note(1): Between 2004 and 2006, Roche manufacturer transferred to LAFEPE manufacturer

in Brazil the process technology and ownership of benznidazole 100 mg product, cancelling

at this time its product registration in Brazil.

6. International availability - sources, if possible manufacturers and trade names

Availability of Pediatric Benznidazole 12.5mg

Pediatric benznidazole (tablet 12.5mg) is produced by Brazilian Pharmaceutical Laboratory of

the State of Pernambuco (LAFEPE, in Portuguese).

One box contains 24 blisters and each blister contains 10 scored tablets. Thus, the box

contains 240 tablets, the equivalent for a treatment.

There is no brand name for the product and it is supplied with a package “LAFEPE

Benznidazol – 12.5mg”.

The product was issued marketing authorization by the Brazilian Drug Regulatory Authority

(ANVISA) in December 2011 (ANVISA registration number: 1018301450062 – see Appendix

5). The product’s registration was granted in Brazil with a shelf life of 24 months.

There are package inserts for both patient and health professionals (Appendix 6 and 7,

respectively) and secondary package are available in Portuguese and Spanish (Figure 1).

Characteristics: tablets containing 12.5mg of Benznidazole

Page 8: proposal for the inclusion of benznidazol pediatric dosage

8

8

Storage conditions: The product should be stored at room temperature of 15 to 30° C,

sheltered from light and humidity.

Figure 1– Secondary package of Lafepe benznidazole 12.5mg (Spanish and Portuguese)

Any interested country – international organizations, control programs, non-governmental

organizations or private sector – can purchase from LAFEPE.

As a contribution to facilitate a sustainable access and international availability, a

Procurement guide in English, Spanish and Portuguese was elaborated with the step-by-step

process to be taken, available LAFEPE’s website:

http://www.lafepe.pe.gov.br/LAFEPE/noticias/noticiario/guiabenznidazol.htm

7. Whether listing is requested as an individual medicine or as an example of a therapeutic group

It is requested that Benznidazole 12.5mg be listed in the WHO Model List of Essential

Medicines for Children, for the treatment of infection due to Trypanozoma cruzi (Chagas

Disease) as an additional dosage form.

Benznidazole tablets 100mg is already listed in section 6.5.5.2 (in both, adult and Children

WHO Model List) as an essential medicine for American trypanosomiasis (Chagas disease).

8. Information supporting the public health relevance (epidemiological information on disease burden, assessment of current use, target population)

8.1 Epidemiological information on disease burden

Chagas disease is caused by the Trypanosoma cruzi (T. cruzi) kinetoplastid parasite and is

endemic in 21 countries of Central and South America, with an estimated 100 million people

at risk of being infected by the parasite3. The main mode of transmission (>80%) is through

triatomine vector, insects known as ‘kissing bugs’ (barbeiro in Brazil, vinchuca in Spanish,

Page 9: proposal for the inclusion of benznidazol pediatric dosage

9

9

etc). Other forms of transmission are blood transfusions and organ transplants (~15%),

congenital/vertical transmission (4%), oral transmission (<1%) and accidental transmission

(<1%)4.

This disease appears in two clinical phases: an acute phase, usually asymptomatic or

characterized by non-specific symptoms that, if left untreated, might progress to a chronic

phase, where 20% to 30% of infected patients develop severe forms of cardiopathy or

digestive megaformations (megacolon or megaesophagus). According to estimates

published by the World Health Organization (WHO) in 2000, there were 18 million people

infected in the Americas, of who some 5.4 million progress to severe cardiopathies and

900,000 to digestive megaformations5. More recent estimates issued by the Pan American

Health Organization (PAHO) indicate that 7.7 million people were infected in 21 countries in

Central and South America in 2005.3

A recent report published by WHO on Neglected Tropical Diseases in 20106 estimates that 10

million people worldwide are infected by T. cruzi, mostly in the endemic areas of 21 Latin

American countries, but also including non-endemic countries such as in the Region of the

Americas (Canada and the United States of America), the Western Pacifi c Region (mainly

Australia and Japan) and the European Region (mainly in Belgium, France, Italy, Spain,

Switzerland and the United Kingdom, but also in Austria, Croatia, Denmark, Germany,

Luxembourg, the Netherlands, Norway, Portugal, Romania and Sweden) as a consequence of

population mobility (see Figure 2).

The morbidity resulting from the chronic phase generates serious social and economic

impacts, causing unemployment and a reduction in productive capability. In Brazil alone, it

is estimated that more than US$ 1.3 billion in wages and industrial productivity were lost

through workers with Chagas disease.7

Page 10: proposal for the inclusion of benznidazol pediatric dosage

10

10

Figure 2: Distribution of cases of Trypanosoma cruzi infection, based on official estimates and status of vector transmission, worldwide, 2006–2009

Source: WHO, 2010.

A recent publication estimates the number of immigrants infected by T. cruzi at around 39,000 in 2003, and estimated the European countries8. In Spain of some 400,000 immigrants living there in 2003 some 12,000 were infected T. cruzi.9 Up to 2007, the number of immigrants from Latin American reached 1,600,000 10 and, if the prevalence of infection remains similar, some 40,000 of these immigrants will be infected by T. cruzi in Spain alone11, as shown in Table 2.

Page 11: proposal for the inclusion of benznidazol pediatric dosage

11

11

Table 2: Estimated number of infected immigrants in Spain, based on immigrant population by country of origin and seroprevalence estimates issued by the Pan American Health Organization (2006).

Based on published seroprevalence estimates, a paper by Bern & Montgomery12 calculates

that, among 300,167 immigrants infected with T. cruzi living in the USA, there are 30,000 to

45,000 cases of cardiomyopathies and between 63 and 315 cases of congenital infections a

year, as shown in Table 3.

Table 3. Calculated prevalence of infection with T. cruzi among people from Latin American living in the USA in 2005.

Country of origin

Immigrant population living in the USA

Prevalence of T. cruzi in countries of origin, %

Estimated Nº of immigrants infected with T. cruzi in the USA

Mexico 16,963,851 1.03 174,388

El Salvador 1,458,014 3.37 49,164

Guatemala 1,014,669 1.98 20,131

Honduras 567,002 3.05 17,311

Argentina 223,931 4.13 9,246

Ecuador 345,204 1.74 6,003

Colombia 554,821 0.96 5,304

Brazil 501,036 1.02 5,106

Bolivia 61,453 6.75 4,149

Nicaragua 223,931 1.14 2,553

Peru 371,980 0.69 2,552

Venezuela 151,350 1.16 1,754

Chile 92,761 0.99 914

Costa Rica 95,761 0.53 509

Paraguay 16,707 2.54 425

Uruguay 51,737 0.66 339

Belize 42,130 0.74 312

Panama 107,601 0.01 6

Page 12: proposal for the inclusion of benznidazol pediatric dosage

12

12

Country of origin

Immigrant population living in the USA

Prevalence of T. cruzi in countries of origin, %

Estimated Nº of immigrants infected with T. cruzi in the USA

Total 22,843,939 1.31 300,167

In Latin America, successful vector control and transfusion transmission reduction programs

run by governments in endemic countries have reshaped the epidemiology of the disease,

resulting in a significant drop in the appearance of new cases, as shown in Table 4, as well as

halting vector transmission in some of the Latin America countries (vector and transfusion

transmission in Uruguay in 1997; vector transmission in Chile in 1999; vector transmission by

through the main domestic vector, Triatoma infestans, in Brazil in 2006; vector transmission

in four provinces in Argentina in 2001 and in one department in Paraguay in 2002).

Table 4: Evolution of some epidemiological parameters for Chagas disease between 1990 and 2000 13

Epidemiological Parameters 1990 2000

Nº deaths x year > 45,000 21,000

Nº cases of human infection 16-18 million 18 million

Nº new cases x year 700,000 200,000

Source: WHO, 2000

In 2001, the estimates for the number of people infected were revised to 9.8 million14, with

the WHO estimating a reduction in the number of deaths caused by thus disease to

13,000.15,16

Page 13: proposal for the inclusion of benznidazol pediatric dosage

13

13

An estimate published by Pan American Health Organization (PAHO) in 2006, indicates a

prevalence of approximately eight million infected individuals and an incidence of 55,185

new cases each year, among which 14,385 are congenital infections17 (tables 5 and 6).

Table 5: Estimated prevalence and incidence of Chagas disease in Latin America, 2005

Countries Nº of people infected (Million)

New cases x year

Total Population

(Million)

Exposed Population

(Million)

Vector Congenital

21 7.5 40,800 14,385

526,95 95,595 55,185

Table 6: Countries affected by Chagas disease

Countries Nº of people

infected (Million)

New cases x year

Total Population (Million)

Exposed Population (Million)

Vector Congenital

Southern Cone Initiative

8. Argentina 1.6 1,300 1,800 39 7.3

9. Bolivia 0.620 10,300 1,500 9.1 3.2229

10. Brazil 1.9 0 5,000 186.8 21.8

11. Chile 0.1602 0 445 16.4 0.8011

12. Paraguay 0.150 900 600 6.3 3.444

13. Uruguay 0.0217 0 20 3.3 0.625

4.4519 12,500 9,365 260.9 37.193

Andean Initiative

14. Colombia 0.436 5,250 1,000 46.8 4,792

15. Ecuador 0.230 2,350 800 13.3 6.2

16. Peru 0.192 3,100 200 28.4 3.455

17. Venezuela 0.310 1,400 600 27 4.944

1,168 12,100 2,600 115.5 19,391

Central American countries and Belize Initiatives

18. Belize 0.002 20 10 0.3 0.1351

19. Costa Rica 0.023 30 60 4.3 1

20. Salvador 0.232 2,500 230 7 2.7

21. Guatemala 0.250 2,200 400 13 2.1

22. Honduras 0.220 2,800 450 7.4 3.5134

23. Nicaragua 0.0586 750 100 5.6 1.2855

24. Panama 0.021 200 50 3.3 1

25. F. Guiana 0.018 400 20

0.15 0.777

26. Suriname 0.5

Page 14: proposal for the inclusion of benznidazol pediatric dosage

14

14

Countries Nº of people

infected (Million)

New cases x year

Total Population (Million)

Exposed Population (Million)

Vector Congenital

27. Guiana 0.7

0.8246 8,500 1,320 42.25 12,511

Other countries

28. Mexico 1.1 7,700 1,100 108.3 26,.5

Source: PAHO, 2006

Regardless of the success attained through vector control programs, vertical transmission

remains an important form of transmission. With an estimated prevalence of T. cruzi

infection among pregnant women varying from 5% to 40%, depending on the geographical

area, this may reach 81% in some rural areas.18,19 With a vertical transmission rate estimated

at around 5%,20,21,22, varying from 1% to 12%,23 congenital Chagas disease will constitute a

major public health problem for many years to come, not only in the endemic countries but

also in countries absorbing significant population flows, until efficacious new treatment

options can prevent the progression of the disease or its vertical transmission.

A retrospective study of mother-to-child transmission of Chagas infections among 278

patients in the chronic phase, born in different Brazilian States to 145 mothers testing

seropositive for T. cruzi, Rassi et al. (2004) estimated the vertical transmission rate at 0.7%

(2/278 children or 2/289 births), consequently not distinguishing between congenital

transmission and that acquired through breast milk24.

Recent data obtained through a serological screening study conducted among children less

than five years old in Brazil, Luquetti et al. (2005) recorded anti T. cruzi antibodies in

nineteen of 9,556 of them (preliminary data for Minas Gerais State).25 These data indicate a

lower prevalence of congenital infections in Brazil, compared to other countries in Latin

America.

8.2 Assessment of current use

Benznidazole (Bz) and Nifurtimox (Nx) are the only two medicines currently available for the

etiological treatment of Chagas disease, and both of them have been used for years.

Benznidazole constitutes the drug of first choice in most of the Latin American countries due

to its greater tolerability and easier accessibility. Nx is usually recommended as an

alternative for cases of intolerance to benznidazole, as its availability has varied over the

years, in addition somewhat less data with regards to its safety and tolerability profile.

The treatment of Chagas disease is recommended for all cases of acute, congenital and

reactive infections among all children, and for patients up to 18 years old in the

indeterminate chronic phase (PAHO, 1998; WHO, 2002). As most infections occur during

childhood, most cases involve children, including newborns diagnosed at birth.

Page 15: proposal for the inclusion of benznidazol pediatric dosage

15

15

Recommendations arise from clinical experiences and studies with patients in the acute

phase and with congenital disease, who have seen that the treatment that reduces the

severity of the treatment, while slowing clinical progression and shortening the duration of

detectable parasitemia.26,27,28,29,30

During the 1970s, several treatment experiments were published with Benznidazole in

children with acute and congenital infections (Barclay et al.31, 1978; Russomando et al.,32,33,

1998, 2005; Cançado et al., 200234; Torrico et al.,35 2004; Salas et al.36,, 2007; Chippaux et

al.,37 Instituto de Recherche pour le Développement [IRD], 2008-2009) and in the

indeterminate chronic phase (de Andrade et al. 38, 1996; Sosa Estani et al. 39,40, 1998, 2002;

Flores-Chavez et al.41, 2006, Duffy et al.42,2009), or comparing the efficacy and tolerability of

benznidazole and nifurtimox in the same age brackets (Blanco et al.43, 2000; Silveira et al.44,

2000), in different age bracket groups (Ferreira, 12,13, 1988, 1990; Streiger et al.45, 2004) as

well as for different age groups and stages of the disease (Schijman et al.4, 2003).

Parasitological responses occur in 60% to 85% of patients during the acute phase, and in

more than 90% of children with congenital infections treated during the first year of life. 46,47 Further information and detail about the clinical evidence will be discussed in Section 10

(identification of clinical evidence).

In Brazil, the Health Surveillance Bureau under the Ministry of Health recommends

“treatment for children and young adults in the chronic indeterminate form and the mild

cardiac and digestive forms.”48

In 2010, during the World Health Assembly it was approved a Resolution “Chagas disease:

control and elimination (WHA 63.20)49” which urges WHO Members States “to promote the

development of public health measures in disease-endemic and non-endemic countries,

with special focus on endemic areas, for the prevention of transmission through blood

transfusion and organ transplantation, early diagnosis of congenital transmission and

management of cases” (paragraph 8).

In 2011, a summary of the recommendations from the Technical Group IVa ‘‘Prevention and

Control of Congenital Transmission and Case Management of Congenital Infections” of the

WHO’s Programme on Control of Chagas disease was made public50.

With regards to the treatment of neonates and infants, both benznidazole and nifurtimox

are recommended for the treatment of congenital cases. Doses for benznidazole are

respectively from 5-7mg/kg/day and up to 10mg/kg/day for infants and infants/neonates by

1 year old, divided in 2-3 doses per day, during 60 days and not less than 30 days. The WHO

recommendation (2011) supports the need of a dosage form of 12.5mg of benznidazol to

facilitate the preparation of pediatric suspension.

Recent case reports of congenital Chagas disease in non-endemic countries, such as

Switzerland and United States of America, have been published51,52, as well as proposal of

screening programme for congenital transmission, such as in Spain 53.

In Switzerland, Jackson et al. (2009) reported two congenital cases, in 2001 and 2006, of

women from Bolivia who delivered their babies in the Geneva University Hospital.

Page 16: proposal for the inclusion of benznidazol pediatric dosage

16

16

Congenital cases were confirmed by positive blood miscroscope examination and PCR for the

infants and positive serologic and PCR for the mothers. Newborns were treat with nifurtimox

(10mg/kg/day) during 60 days, without notable adverse effects. Parasitemia became

undetectable at the end of treatment and at subsequent serologic tests.

The authors also developed a retrospective serologic survey for T. cruzi infection with serum

samples stored in the Hospital from 72 pregnant women from Latin America countries who

received care in the Hospital and 9,7% were positive for T. cruzi.

In 2008, the Geneva University Hospitals set up a systematic Chagas disease screening of

pregnant women at risk (those from Mexico, Central and South America) and newborns. For

positive diagnosis, all women (after completion of breastfeeding), newborns and siblings are

offered treatment.

More recently, the first case report of congenital Chagas in United States of America (USA)

was published by CDC in July 2012. In August 2010 a boy was born to a mother who moved

to US from Bolivia. Physicians learned from the mother that at her previous pregnancy she

had been told to have Chagas disease. The child was diagnosed positive by identification of

T. cruzi trypomastigotes in the blood smear, and both serologic tests for anti-T. cruzi

antibodies and PCR were positive. The baby was treated with benznidazole during a 60 days.

Follow-up tests at age of 10 months showed the baby was cured based negative results of T.

cruzi PCR and serologic tests.

Basile et al. (2011) described the screening programme for congenital transmission of

Chagas disease, implemented from January 2010, in Catalonia (Spain) developed with the

WHO Department of Neglected Tropical Disease. The diagnosis for Chagas was offered in

the 1st semester of pregnancy following PAHO recommendations of two serological tests. For

serological positive mothers, newborns are diagnostic screened and in case of positive

parasitological test at birth or a positive serological result at nine months, it is adopted the

treatment, having benznidazole more widely used. Siblings, if needed, are also treated.

Despite widely accepted consensus on the efficacy of treatment for children and its greater

tolerability compared to administration in adults (described in next topics) and the

treatment recommendations for the pediatric population, pediatric formulations of these

drugs used were until recently not available.

Benznidazole was produced by the Pharmaceutical Laboratories Roche from 1971 onwards,

and the technology was transferred to a Brazilian manufacturer in 2003: the Laboratorio

Farmacêutico do Estado de Pernambuco S/A LAFEPE. Benznidazole was registered in 2006

as a new medicine with ANVISA (LAFEPE BENZNIDAZOL® 25351.111801/2006-44 11/2011,

Resolution RE Nº 3,732, dated November 17, 2006, published in the Federal Government

Gazette on November 20, 2006), with registration renewed in 2009 (Registration Nº

101830145, published on March 9, 2009 expiring in November 2011) and in 2011 (published

in Resolution 5,720, 16 December 2011).

Produced as 100 mg tablets, benznidazole is administered twice a day, at doses of 5-7 mg/Kg

for adults during 30-60 days and 5-10 mg/Kg for children during 60 days (Appendix 6 and 7).

Page 17: proposal for the inclusion of benznidazol pediatric dosage

17

17

Consequently, for treatment of most children, the tablet must be cut into two, four or more

fractions, which are administered directly or crushed and diluted in liquid, presenting a risk

of possible overdose and increased toxicity, particularly for smaller or undernourished

children, or else under-dosing with a possible loss of efficacy. Figure 3 illustrates the most

frequent ways of administering benznidazole to children.

Figure 3 – Forms of administration of Benznidazole, 100 mg tablets to children

There are no studies on the stability of these different extemporaneous forms of

administration. Extemporaneous preparations undertaken by healthcare workers or

mothers have been shown to be imprecise, as shown in Graph 1. The weight of a split tablet

may vary between 50% and 150% of the real weight of half a tablet.54

Crushed

Tablets

Re-suspension in

10 mL of water

Volume equivalent to

the dose is measured in

the syringe

Administration

by mouth

Administration

by mouth

Powder with equivalent dose is weighed

analytically = capsules

Capsule content

reconstituted

Page 18: proposal for the inclusion of benznidazol pediatric dosage

18

18

Page 19: proposal for the inclusion of benznidazol pediatric dosage

19

19

8.2.1 Target population

The target population for the benznidazole 12.5 mg pediatric dosage form is children up to

two years old, mostly congenital cases. The rational to establish the dose and the age range

are described in the next paragraphs.

Legislators and physicians have long emphasized the pressing need for a pediatric dosage

form for the treatment of Chagas disease, particularly the TDR/WHO Scientific Working

Group for Chagas Disease (2005) and the TDR/WHO Working Group on Chagas Disease

(2007), which highlighted the unmet need for a pediatric dosage form for the treatment of

Chagas disease.

Thus, in July 2008, DNDi and the Pernambuco State Pharmaceutical Laboratory (Laboratorio

Farmacêutico do Estado de Pernambuco - LAFEPE) established a partnership designed to

bridge this gap, implementing a project developing a pediatric dosage form of benznidazole.

The characteristics of the pediatric dosage form were established through a review of the

current treatment recommendations; a compilation of the databases on children treated in

Latin America, and a comparison of the current doses prescribed, in order to determine the

target population for the pediatric dosage form and the therapeutic margins (upper and

lower limits), in addition to a definition of the pharmaceutical form and composition

established by a specialist panel.

Table 7 summarizes the recommended dosages identified through a systematic review of

the WHO Guides, National Control Programs and medical literature.

Table 7 - Recommended dosage of Benznidazole for Chagas disease infections

Source Age Range

WHO – Consensus - Specialist Group Chagas Disease Control Experts

Congenital infections: 5-10 mg/kg/day

WHO – Prescription model Children >= 12 y.o.:5-7 mg/kg/day, Children <12 y.o.:10 mg/kg/day (interval not stated)

Package Insert - Hoffman-La Roche Children >= 12 y.o.:5-7 mg/kg/day, Children <12 y.o.:10 mg/kg/day (interval not stated)

Package Insert - Roche. Radanil® 5-8 mg/kg/day 2x/day for 60 days

Package Insert - Roche. Rochagan® 5-7 mg/kg/day 2x/day by mouth for 30-60 days Children < 12 y.o., especially with acute infections: up to 10 mg/kg/day for the first 10-20 days of treatment

Ministry of Health, Health Surveillance Bureau.

Adults: 5 mg/kg/day, Children: 5-10 mg/kg/day two or 3x/day by mouth for 60 days

Page 20: proposal for the inclusion of benznidazol pediatric dosage

20

20

Source Age Range

Brazilian Ministry of Health, Health Surveillance Bureau.

Acute phase, congenital infections, immunocompromised and transplant patients: 8 mg/kg/day 2x/day by mouth for 60 days

Mazza Chagas Disease Council, Argentine Cardiology Society

5 mg/kg/day for 30-60 days

The recommended benznidazole dosages indicate variations in milligrams by kilogram of

weight in the recommended dose-category. In general, the doses recommended for

children are higher than those for adults. The main variations appears between the

recommended dosage of 5-10 mg/kg, where 10 mg/kg is a maximum acceptable dose, with

recommendations of 10 mg/kg as the target dose, with no mention of any acceptable margin

or error for this dosage. No recommendation offers guidance on the practical aspects and

challenges of administering accurate doses to children, with the 100 mg formulation

currently available.

In order to confirm the therapeutic margins based on clinical experience and identify the

pediatric population for which the administration of the correct dose constitutes the

greatest problem, and which would benefit the most from a pediatric dosage form,

treatment data were compiled for patients with Chagas disease in Latin America, with

various specialists in treating pediatric T. cruzi infections being contacted.

Secondary analyses of anonymous treatment data were also conducted on the basis of 2,769

records drawn from ten sources, supplied by the contacted congenital Chagas experts in

different countries. Figures 4 and 5 present the age distribution of the compiled patient

population.

Page 21: proposal for the inclusion of benznidazol pediatric dosage

21

21

Figure 4 – Patient distribution by age (based on compiled data, n = 2,779)

Figure 5 - Age distribution (months) among children less than one year old (n = 247).

Although not aiming to constitute a representative sample of patients with Chagas disease in

Latin America, these compiled data offer an overview of children population treated by age

range for which dosing data was available, indicating the existence of three specific groups

of pediatric patients: congenital infections confirmed by microscopy, for which treatment is

initiated at birth, congenital infections diagnosed through serology during the second half of

Page 22: proposal for the inclusion of benznidazol pediatric dosage

22

22

childhood; and early indeterminate infection among schoolchildren, detected more

systematically through Chagas disease screening programs run in schools.

Considering the therapeutic margins currently in place and the weight-age distribution in

this data compilation (Figure 6a and 6b), it is clear the significant variation of dosing the

adult tablet of 100 mg.

Figure 6a and 6b – weight-age distribution

When analyzing the dose prescribed in mg/kg in this data compilation, it is apparent that the

daily dose is maintained for a therapeutic interval of 5-10 mg/kg/day for most of the children

(Figures 7a and 7b), except for infants under one year old. In this group, the doses vary from

5 mg/kg to 15 mg/kg, while for new-borns, a significant proportion children receive doses

exceeding 10 mg/kg, reflecting the difficulties associated with fragmenting the 100 mg

tablet. Consequently, the group that would benefit most with a pediatric dosage form

consists of infants up to a year old with congenital infections.

Figure 7a - Dose by age bracket. All age

brackets (n=2,424)

Figure 7b - Dose by age bracket. Dose

administered at <1 year (n=317)

0.0

10.0

20.0

30.0

40.0

50.0

60.0

70.0

80.0

90.0

100.0

110.0

0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20

We

igth

(K

g)

Age (months)

----Median age = 0.15 months

0.0

1.0

2.0

3.0

4.0

5.0

6.0

7.0

8.0

9.0

10.0

11.0

12.0

13.0

14.0

15.0

0 1 2 3 4 5 6 7 8 9 10 11 12

We

igth

(K

g)

Age (months)

----Median age = 0.15 months

0

2

4

6

8

10

12

14

16

18

20

22

0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20

Do

se

(m

g/k

g)

Age (years)

0

2

4

6

8

10

12

14

16

18

20

22

0 1 2 3 4 5 6 7 8 9 10 11 12

Do

se

(m

g/k

g)

Age (months)

Page 23: proposal for the inclusion of benznidazol pediatric dosage

23

23

After an analysis of the current benznidazole dose recommendations and the doses

prescribed in clinical practice, a panel of specialists1 defined a therapeutic interval for the

pediatric dosage form of 5-10 mg/kg, twice a day for 60 days for children under 12 years of

age, consistent with the recommendations issued by the Chagas Disease Control Technical

Specialists Group with the WHO (2002).55 This panel of experts indicated that, despite the

lack of pharmacokinetic data available for support in this therapeutic range, such as the ideal

range for children, benznidazole seems to be tolerated better by small children than by

adults31, for whom toxicity is a significant problem. Furthermore, the treatment is more

efficacious among small children, reaching parasitological clearance rates of more than 90%

for congenital infections treated during the first year of life, as mentioned and explained in

further detail in the section on clinical efficacy overview.

The recommended interval of administration at twice a day is based on the benznidazole

terminal half-life, which varies from 10.5 to 13.6 hours, with an average of 12 hours56,57.

With regard to the definition of the pharmaceutical form, after considering the current

forms of administration for benznidazole (see Figure 3) and the recommendations in the

international guidelines, particularly the Treatment Guidelines for Scaling up Antiretroviral

Therapy in Resource-limited Settings58 issued by the WHO which “strongly encourages the

development of formulations appropriate for pediatric use, particularly solid forms that may

be taken by pediatric patients (for example, dissolvable tablets or capsules that can be

opened), as the conservation times for liquid formulations may be shorter than those of the

solid formulations, in addition to being more expensive and harder to store, and possibly

requiring the use of syringes for correct administration”. Consequently, the panel of experts

agreed on the decision to develop a solid pediatric presentation of benznidazole in the form

of a rapidly disintegrating tablet, as this would allow the use of a minimal quantity of non-

toxic excipients, as well as being easy to produce, more stable and cheaper than the liquid

form, for easy, reliable administration, particularly for long-duration home treatment.

For definition of tablet strength, an acceptable therapeutic interval of 5 to 10 mg/kg was

agreed, with the ideal administration consisting of 1-2 tablets orally, with a maximum of

single fractionation.

The adult tablet of 100 mg is designed for patients weighing more than 20 kg (see Figure 8),

which constitutes the group of children at school age, greater than eight years old and with

early indeterminate infection, while half the adult tablet (or 50 mg tablet) could encompass

the weight range between 10 and 20kg.

1 Chagas disease Expert Panel Dr. Jaime Altcheh, Hospital de Niños Ricardo Gutierrez, Buenos Aires, Argentina; Dr. Laurent Brutus, Institut de Recherche pour le Développement, La Paz, Bolivia ; Dr. Sérgio Sosa Estani, Centro Nacional de Diagnóstico e Investigación de Endemo-epidemias (CeNDIE) ANLIS Dr. Carlos G. Malbrán, Ministry of Heath, Argentina

Page 24: proposal for the inclusion of benznidazol pediatric dosage

24

24

Figure 8: Range of the adult tablet (100 mg)

The strength of 12.5 mg was selected for the pediatric tablet, equivalent to 1:8 of the adult

tablet of 100 mg, in order to reduce the need for splitting the tablet for administration to

new-borns. The 12.5 mg pediatric tablet thus encompasses a broad range of age and weight

for small children, as shown in Figure 9.

0

5

10

15

20

25

30

5 10 15 20 25 30 35 40 45 50 55 60 65 70 75 80 85 90 95 100

Be

nzn

ida

zo

le d

ose

(m

g/k

g)

weight (kg)

Assessed tablet strength =100mg

1 tab AM + 1 tab PM

2 tabs AM + 2 tabs PM

3 tabs AM + 3 tabs PM

4 tabs AM + 4 tabs PM

lower

upper dose range cut-of f

T1_lower

T1_upper

T2_lower

T2_upper

T3_lower

T3_upper

T4_lower

T4_upper

Figure.

Assumes 1) equal split of the daily doses AM + PM: 1 tab = 1tab AM + 1tab PM2) use of full tablets only, 3) PK linear across all age groups.

Page 25: proposal for the inclusion of benznidazol pediatric dosage

25

25

Figure 9: Range of the pediatric table (12.5 mg)

As benznidazole is a medication whose use is established for adults as well as the pediatric

population, and has already been registered under international standards (Radanil®,

Rochagan®) and in Brazil (LAFEPE Benznidazole®) for the treatment of Chagas disease, the

development plan focused on the registration requirements for the inclusion of a new

pediatric presentation and indication. The development strategy complied with the latest

Brazilian domestic and international guidelines, especially Executive Board Resolution RDC

No. 48/09 - on Post-Registration of Medication59.

The main elements taken into consideration in this process included the following points:

The well-established therapeutic indication of benznidazole as the first line treatment

for Chagas disease in Brazil, and in most of the endemic countries;

The inclusion of benznidazole in the list of essential medications drawn up by the

WHO and in Brazil's Essential Medications Listing (Rename)60, as well as its inclusion

in the list of strategic products established by Edict Nº 978 promulgated on May 16,

2008, replaced by Edict Nº 1,284 on May 26, 2010.61

The existence of a therapeutic consensus (WHO, PAHO and assorted national Chagas

disease control programs) regarding the recommendation for treatment of all acute

and congenital infections, and reactivation for all children;

The balance of evidence confirming the efficacy of benznidazole treatment for

children;

0

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15

Be

nzn

ida

zo

le d

ose

(m

g/k

g)

weight (kg)

Assessed tablet strength =12.5mg, therapeutic dose range = 5 to 10 mg/kg

0.5tab AM + 0.5tabPM

1tab AM + 1tab PM

2tabs AM +2 tabs PM

3tabs AM + 3 tabs PM

4tabs AM + 4 tabs PM

lower

upper dose range cut-of f

T0.5_lower

T0.5_upper

T1_lower

T1_upper

T2_lower

T2_upper

T3_upper

T3_lower

T4_upper

T4_lower

Figure.

Assumes 1) equal split of the daily doses AM + PM: 1 tab = 1tab AM + 1tab PM2) use of half tab only in those < 5kg, rest full tablets only, 3) PK linear across all age groups.

Page 26: proposal for the inclusion of benznidazol pediatric dosage

26

26

The availability of safety and tolerability data, indicating that benznidazole is better

tolerated among the pediatric population than by adults;

The composition of the pediatric presentation with the same qualitative composition

as the adult formulation and comparative dissolution profile data for both

presentations; and

The maintenance of the dosage schedule recommended by the Brazilian Ministry of

Health and WHO (5 – 10 mg/kg/day administered twice a day for 60 days).

As a result of the established use of benznidazole for the pediatric population, it was not

considered necessary to conduct in vivo clinical trials in order to support the pediatric

dosage form registration dossier. Following registration, a population pharmacokinetics

study in children aged 0 to 12 years of age has been recently concluded and provides

additional data in support to the efficacy and safety of the benznidazole 12.5 mg and 100 mg

tablets in the treatment of Chagas disease.

In Brazil, benznidazole pediatric dosage form (tablet 12.5mg) is already included in the

Brazilian Essential Medicines List (RENAME) 201262.

9. Treatment details (dosage regimen, duration; reference to existing WHO and other clinical guidelines; need for special diagnostics, treatment or monitoring facilities and skills)

9.1 Dosage regimen, duration

Benznidazole 12.5mg (oral tablets): 5-10mg/kg/day, twice a day, during 60 days.

Table 8 – Summary of the categories of weight per dose for tablets Benznidazole 12.5mg

Weight (Kg) Recommended Dose (5 – 10 mg/kg)

2,5 a < 5 kg 1 tablet in twice a day during 60 days (total dose of 25 mg per day)

5 a < 10 kg 2 tablets (25 mg) in twice a day during 60 days (total dose of 50 mg per day)

10 a < 15 kg 3 tablets (37,5 mg) in twice a day during 60 days (total dose of 75 mg per day)

Source: Lafepe benznidazole package insert – Appendix 6.

9.2 Reference to existing WHO and other clinical guidelines

Page 27: proposal for the inclusion of benznidazol pediatric dosage

27

27

According to the Second Report of the WHO Expert Committee (2002)63, nifurtimox (a

nitrofuran derivative) and benznidazole (a nitroimidazole) are the only medicines used for

the treatment of the acute phase of Chagas disease and for congenital infection.

In congenital cases, recommended treatment of full-term neonates is with a daily dose of

10mg/kg of benznidazole. In this report, suggested treatment must start with a dose of

5mg/kg daily and if, after 3 days of treatment, there is neither leukopenia nor

thrombocytopenia, the dose should be increased to 10mg/kg daily.

As discussed in section 8.2 (Assessment of Current Use), in 2011 WHO updated its

recommendation for the diagnosis, treatment and control of newborns, siblings and

pregnant women related to congenital Chagas disease48. A summary of the

recommendations from the Technical Group IVa ‘‘Prevention and Control of Congenital

Transmission and Case Management of Congenital Infections” of the WHO’s Programme on

Control of Chagas disease was made public. With regards to the treatment of neonates and

infants, both benznidazole and nifurtimox are recommended for the treatment of congenital

cases. Doses for benznidazole are respectively from 5-7mg/kg/day and up to 10mg/kg/day

for infants and infants/neonates by 1 year old, divided in 2-3 doses per day, during 60 days

and not less than 30 days. In the WHO recommendation (2011) there is a clear support to

the need of a dosage form of 12.5mg of benznidazole to facilitate the preparation of

pediatric suspension.

In 2012, the WHO Drug Information also published a reference with regards to the

benznidazole pediatric 12.5mg64.

9.3 Need for special diagnostic or treatment facilities and skills

In most endemic countries, Chagas disease diagnosis and treatment is organized through

vertical programs that ensure the necessary specific training and equipment required. In

comparison with current practice, no additional special diagnostic or treatment facilities are

needed.

10. Summary of comparative effectiveness in a variety of clinical settings:

There are no comparative studies on the effectiveness in a variety of clinical settings. The

following sub-sections will provide an overview of the clinical evidence on the efficacy of

benznidazole in children.

The rationale behind the development of the benznidazole dosage form 12.5mg considered

the fact that the target pediatric population is very small, thus clinical studies to provide

information on comparative effectiveness would take severeal years to achieve the

appropriate sample size. In the risk benefit assessment, this would mean additional years in

Page 28: proposal for the inclusion of benznidazol pediatric dosage

28

28

which children would be treated with benznidazole adult formulation with a significant

variation in the dose administrated, as discussed previously.

In addition to the existing clinical evidence in support to the efficacy and safety of

benznidazole in children, DNDi in collaboration with partners also implemented a population

pharmacokinetic study for evaluation of the pediatric dosage form 12.5mg (section 11.7).

10.1 Identification of clinical evidence and summary of available data

10.1.1 Studies of Benznidazole efficacy in congenital infections

A number of studies have been conducted in children with congenital infections, treated

with benznidazole.

A randomized clinical study was conducted by the IRD (2008-2009) on T. cruzi transmission,

comparing the reduction in anti T. cruzi antibodies titers among non-infected new-borns,

with those of new-borns with congenital infections treated with two therapeutic

benznidazole schemes, in order to determine the serological cure criteria. The other

available studies were conducted in situations of pre-natal screening , upon detection of

congenital infections among newborns of mothers infected by T. cruzi, and following

determination of the prevalence of a congenital transmission (Blanco et al., 2000;

Russomando et al., 1998; Salas et al., 2007; Torrico et al., 2004) or evaluating the PCR assay

as a tool for diagnosing and assessing parasitological response to treatment (Schijman et al.,

2003).

Although main purpose of these studies was not to assess the efficacy of benznidazole, these

studies provide evidence of the efficacy of treating congenital infections, as described below

and summarized in Table 9.65

In a randomized controlled study conducted by IRD (2008-2009) in Bolivia of 111 new-borns

with congenital infections diagnosed at birth through direct microscopic observation

(microhematocrit [MH]), 58 children were treated from the first day of life with benznidazole

2.5 mg/kg/day administered twice a day for 60 days, and 52 children with benznidazole 7.5

mg/kg/day administered once a day for 30 days. In order to compare the reduction in anti T.

cruzi antibody titers, two other study arms included 68 new-borns with seropositive mothers

who did not present parasitemia at birth, and 78 new-borns with seronegative mothers.

Blood samples were taken at 30 and 60 days and 10 ± 2 months and, with seropositive ELISA

testing after ten months, every two months until seroconversion. All parasitology tests (MH)

showed negative results before the ninth month. After ten months of follow-up, ELISA was

negative for 90.7% of the children treated (98/108) with all the seropositive tests becoming

negative after 16 months, indicating a response to treatment. No significant difference was

noted in the proportion of children presenting seroconversion after ten months between the

groups treated with different therapeutic schemes (p=1.0), nor among the average figures

for the antibody titers.

Page 29: proposal for the inclusion of benznidazol pediatric dosage

29

29

In the study conducted by Blanco et al. (2000) in Argentina, 32 children born to infected

mothers and diagnosed as positive up to six months old through parasitology (MH) and

serology tests (IHA, ELISA [IgG and IgM antihuman] and IIF in case of IgG serology findings)

were treated orally with Nx at 10 mg/kg per day, two or three times a day for 60 days (29

children) or with benznidazole, 5 mg/kg per day, two or three times a day for three days (3

children presenting adverse reactions to Nx). The 32 children treated presented negative

parasitology (MH) tests when examined between 6 and 24 months after the end of

treatment, and 30 of the 32 children presented seronegative tests, indicating response to

treatment of congenital infections, regardless of the medication administered.

In the study conducted by Russomando et al. (1998) in Paraguay, six children born from

infected mothers and diagnosed as positive through direct microscopic observation (MH)

and/or hemoculture (Hc), IIF, ELISA and the PCR technique, were treated with benznidazole,

7 mg/kg per day, taken twice a day for 60 days. Treatment began at different times: two

babies were treated at birth, and four others began treatment at between 3 months to 22

months after birth. After 24 months of follow-up, all babies presented negative results in

the parasitology tests (Hc, MH, PCR) with negative seroconversion by IIF and ELISA.

Seroconversion occurred at between two months and eight months after the end of the

treatment, suggesting better outcomes for early treatment.

In the prospective study conducted by Schijman et al. (2003) in Argentina, 40 of the 152

children born to mothers sero-reactive for T. cruzi, diagnosed by MH and PCR (50 children

between 0 and six months old) and through serology (IHA, ELISA) and PCR (102 children

between seven months and 17 years old), were treated with Nx, 10-15 mg/kg/day or

benznidazole, 5-8 mg/kg/day in two doses per day for 60 days. The efficacy of the treatment

was assessed by age group, and the treated children were monitored for 36 months after

the end of the treatment. No differences were observed in clinical and serological results,

nor on the PCR testing between the two treatment groups. 100% of children between 0 to 3

months old age (10 children), and 66.7% of children between seven months to two years old

(6 children) were considered cured based on the serology test (negative for igG anti T. cruzi

antibodies), while for the age group more than 3 years old (24 children), cures were

documented in only 12.5% of the cases during the study observation period (P = 0.023). It

was also noted that seroconversion occurred faster in children who began treatment during

the first few months of life, indicating the possibility of greater efficacy for early treatment,

despite difficulties in determining cures for patients in the indeterminate phase, as

conventional serology remains positive for many years after treatment.

Table 9 summarizes the data on the efficacy of the main clinical studies in children with

congenital infections.

Page 30: proposal for the inclusion of benznidazol pediatric dosage

30

30

Table 9 – Overview of data on the efficacy of the main clinical studies in children with congenital infections

Reference Age (years) Benznidazole Nifurtimox Design* Follow-up (months) Lost in FU (%)

Efficacy Measurements †

n Dose (mg/kg/d)

Time (days)

n Dose (mg/kg/d)

Time (days)

Serological Test (% neg)

Parasitological Test (% pos)

Russomando 1998

Congenital <2

6 7-10 60 - - -

nC nR nB

24m } 0%

(EIA, IIF) } 100%

(Hc,MH,PCR) } 0%

Blanco 2000

Congenital <1

3 -

5 -

30 -

- 29

- 10

- 60

C nR nB

24m } 6%

(EIA,IHA,IIF) } 94%

(MH) } 0%

Schijman 2003

Congenital <2

16 {

5-8 -

60 -

-

- 10-15

- 60

C nR nB

36m } 0%

(EIA,IHA) } 87%

(MH,PCR) } 0%

Chippaux (IRD)

2008 -2009

Congenital <1

(Control=not

infected)

68 59 52

NT 5

7.5

-

60 30

- - -

C R B

10m 0% 2% 4%

(EIA,IC) 100% 91% 90%

(MH a 2m) 0% 0% 0%

Key

Design: C (controlled: control or comparison group), nC (not controlled); R (randomized), nR (not randomized); B (blind), nB (not blind)

† Efficacy Measurements: IHA (Indirect Hemagglutination, IIF (Indirect Immunofluorescence), EIA (Enzyme Immunoassay), IC (Immunochromatography), MH (Microhematocrit), HC (hemoculture), PCR (Polymerase Chain Reaction molecular assay).

Source: IRD 2009

Page 31: proposal for the inclusion of benznidazol pediatric dosage

31

31

10.1.2 Studies of the efficacy of benznidazole in children with acute infection

Table 10 presents an overview of the treatment efficacy data for patients in the acute phase

of benznidazole, in three non-randomized, open-label, controlled studies.

Barclay et al. (1978) followed 139 cases of acute infection during 18 months, mostly

children, diagnosed through xenodiagnosis and the Strout technique. 107 patients were

treated with progressive doses of benznidazole, starting at 3 mg/kg/day until reaching a

dose of 7.5 – 10 mg/kg/day after 12 days, which was then maintained through to the end of

treatment at 30 days; 32 patients received a dose of 5 mg/kg/day during 30 days. The

efficacy of the treatment was assessed in 86 patients through parasitology (xenodiagnosis)

and serology tests (Complement Fixation Test (CFT), IIF and IHA), conducted systematically

during a period of 18 months. At the end of the treatment, the Strout test proved negative

for 86% of the children. At 18 months, the accumulated percentage of negative

xenodiagnoses was 88%, with a negative CFT for 87% of children examined (27/31) and the

IIF presenting negative findings or low titers in 91% of children without parasitemia

examined (41/45; p <0.001).

In the cohort study conducted by Ferreira (1988), a total of 38 children with acute infections

were monitored, with 17 children between 2 and 18 years old treated with oral benznidazole

5 mg/kg/day for 60 days and monitored for 9 years, and 21 children between 6 and 13 years-

old treated with Nx 15 mg/kg/day for 90 days monitored for 15 years. At the end of the

follow-up period, 100% of the patients in both groups presented negative serology tests

(IHA, IIF, CFT) and no patient in either group presented positive parasitology tests

(xenodiagnosis).

Cançado et al. (2002) monitored a group of 21 patients with acute infections between 7

months and 60 years old, for a period of 13 to 21 years, treated with benznidazole between

1974 and 1982, with therapeutic schemes varying from 5 to 10 mg/kg/day, taken two or

three times a day, for periods varying between 30 and 60 days. During follow-up period of

13 years, 16 (76%) of the patients presented negative serology tests (IHA, IIF and ELISA) and

were defined as cured. The other five patients presented persistently positive tests,

indicating treatment failure.

Page 32: proposal for the inclusion of benznidazol pediatric dosage

32

32

Table 10 – Overview of efficacy data for the main clinical studies in children with acute infections

Reference Age (years)

Benznidazole Nifurtimox Design* Follow-up (months) Lost in FU (%)

Efficacy Measurements†

n Dose (mg/kg/d)

Time (days)

n Dose (mg/kg/d)

Time (days)

Serological Test (% neg)

Parasitology Test (% pos)

Barclay 1978

Children

?

107 32

7.5-10

5

30 30

- - -

C nR nB

18 m

} 30%

87% (CFT) 91% (IIF)

12% (xeno)

14% (Strout)

Ferreira 1988

6-13 2-18

-

17

- 5

-

60

21 -

15 -

90 -

C nR nB

15a: 28% 9a: 41%

(IHA,IIF,CFT) 100% 100%

(xeno) 0% 0%

Cançado 2002

<10

11-60

6

15

10-20 5-10

40-60 30-60

- - -

C nR nB

13a

} 0%

(EIA,IHA,IIF)

} 76%

Not used

Key

Design: C (controlled: control or comparison group), nC (not controlled); R (randomized), nR (not randomized); B (blind), nB (not blind)

† Efficacy Measurements: IHA (Indirect Hemagglutination, IIF (indirect immunofluorescence), EIA (Enzyme Immuno Assay), Xeno (Xenodiagnosis),

Strout (Strout Technique) Source: IRD 2009

Page 33: proposal for the inclusion of benznidazol pediatric dosage

33

33

10.1.3 Efficacy studies of benznidazole in children in the indeterminate chronic phase

There are many trials providing evidence of the efficacy of treatment during the indeterminate

chronic phase in children and adolescents, whose findings are presented briefly in Table 11.

This section highlights two randomized controlled double-blind clinical trials, in which patients were

monitored for lengthy periods of time: the study by de Andrade et al. (1996), conducted between

1991 and 1995 in Goiás State, with follow-up for six years, and the study of Sosa Estani et al. (1998),

conducted in Argentina during the same period, with follow-up of nine years.

In the study conducted by Andrade et al. (1996), 129 children from 7 to 12 years old with three

positive serology findings for Chagas (IHA, IIF and ELISA) who were asymptomatic, were randomised

into two groups. The experimental group (64 children) received treatment with benznidazole at 7.5

mg/kg/day for 60 days and the control group (65 children) received a placebo. 88.7% of children (58

treated and 54 of the control group) were monitored for a total period of six years.

The primary efficacy measurement consisted of negative seroconversion of ELISA test (disappearance

of the antibodies directed to a purified trypomastigote glycoconjugate) after three years of follow-up.

Seroconversion occurred in 37 of the 58 children (58%) who completed treatment with benznidazole,

and for three of the 65 children in the control group (5%). The treatment efficacy was estimated at

55.8% (95% CI, 40.8-67.0) for intention to treat and 64% by protocol. At the end of three years of

follow-up, the children who received benznidazole still presented mean IF antibody titers that were 5

times lower than children in the control group (196 [147–256] vs 1068 [809–1408] p<0·00001).

The treatment efficacy after three years was also assessed by Galvão et al.66 (2003) through PCR

assays of 111 children (58 of the experimental group and 53 of the control group) who participated in

the clinical trial conducted by de Andrade et al. At the end of the follow-up period, the PCR assay

was positive for 39% of patients treated with benznidazole and for 64.2 % of patients receiving a

placebo (P= 0.01).

After six years of follow-up, 33 of the 37 children treated with benznidazole and presenting negative

seroconversion after three years remained seronegative, while 14 of the 21 children in this group

refuted a positive serology in this group, resulting in 47 children with negative seroconversion and six

seropositives (cure rate of 88.7%). In the control group of 52 children who were seropositive after

three years of follow-up, 32 remained seropositive after six years, while 11 presented sero-

negativisation (26.1% of negative seroconversion). Consequently, the benznidazole efficacy

estimated by the per-protocol analysis and by intention-to-treat was respectively 84.7% (confidence

interval [CI] of 95% – 66.8-92.9) and 64.7% (confidence interval [CI] of 95% – 50.2-78.7), indicating

the efficacy of benznidazole for children in the early chronic phase.67

In the study conducted by Sosa Estani et al. (1998), 55 children between 6 and 12 years old were

treated with benznidazole 5 mg/kg/day for 60 days and 51 children in the same age range received

placebos. The children were monitored for 48 months and different serology tests (ELISA, IHA, IIF)

were conducted before and at 3, 6,12, 24 and 48 months after the start of treatment. The efficacy of

Page 34: proposal for the inclusion of benznidazol pediatric dosage

34

34

the treatment was assessed through ELISA testing using a T. cruzi flagellate protein (F29). In the

experimental group, the negative seroconversion rate through the ELISA F29 rose from 35.7% to

62.1% after six and 48 months respectively, while seroconversion did not occur in any of the children

in the control group.

Through conventional serology, negative seroconversion occurred in 11.3% (5 out of 44) of the

children in the experimental group (P = 0.05) and in 4.5% (2 out of 44) of the children in the control

group (P= 0.05) after 48 months. Children treated with benznidazole also presented a significant

reduction in the geometric mean IHA and IIF antibody titers, while no significant changes were noted

in the control group. Xenodiagnosis conducted after 48 months of follow-up proved positive in 4.7%

of children treated with benznidazole compared to 51.2% of children of the control group (P = 0.05).

After 9 years (108 months) follow-up, 77% of the treated children presented negative conventional

serology (ELISA, IIF), of whom 88.2% were in the 5-9 age bracket and 69% in the age bracket between

10 and 14 years old age.

Sosa Estani et al. (2002) also accompanied the serological findings by age among two groups of

children between 1 and 14 years treated with benznidazole 5 mg/kg/day during 30 days through the

Argentine Public Health System. In the first group of 147 children treated in 1994, the follow-up at 20

months proved negative for conventional serology (IHA, ELISA) in respectively 49.3%, 32.7% and 0%

of children in the 0 to 4 years, 5 to 9 years old and 10 to 14 years old age range. In the second group

of 40 children treated in 1995, the 5 years follow-up showed negative conventional serology in

respectively 91.7%, 69.2% and 54.5% of children in the 0 to 4 years, 5 to 9 years and 10 to 14 years

age groups.

These findings corroborate the efficacy of treatment with benznidazole of the early indeterminate

form of disease in this age range, suggesting better outcomes for treatment among younger children.

Page 35: proposal for the inclusion of benznidazol pediatric dosage

35

35

Table 11 –Overview of the main clinical trials examining efficacy in children early chronic infection

Reference Age (years)

Benznidazole Nifurtimox Design* Follow-up (months) Lost in FU (%)

Efficacy Measurements†

n Dose (mg/kg/d)

Time (days)

n Dose (mg/kg/d)

Time (days)

Serology Test (% neg)

Parasitology Test (% pos)

Ferreira 1990

<18

50 -

5-8

-

60 -

-

50

-

10-15

-

60-120

C nR nB

24m

} 0%

(CFT,IHA,IIF) 10% 6%

(xeno) 30% 50%

de Andrade 1996; 2004

Galvão 2003

7-12

65 64

P 7.5

60 60

- - -

C R B

PP a‡

36m

17% 9%

72m

29% 17%

(atEIA) 36m 5%

64%

(atEIA) 72m 26% 89%

(PCR 36m)

64% 40%

Sosa Estani 1998; 2002

6-12

51 55

P 5

60 60

- - -

C R B

PP a‡

48m 14% 20%

(F29EIA) 0%

62%

(EIA,IHA) 4%

11% 108m: 77%

(xeno) 51% 5%

Silveira 2000

7-12

7-12

5-7

-

60 -

- 2

-

7-8

-

60-90

C nR nB

8 a 20y } 0%

(CFT,IHA,IIF) 10% 0/2

(xeno) 10% 0/2

(PCR) 10% 0/2

Sosa Estani 2002

1-4 5-9 10-14

83 74 30

5 5 5

30 30 30

- - -

nC nR nB

20m / 60m

} 0%

20m (EIA,IHA) 60m Not used

49% 33% 0%

92% 69% 54%

Schijman 2003

3-17

24 {

5-8

-

60 -

- 10-15

- 60

C nR nB

36m } 0%

(EIA,IHA) } 12%

(PCR) } 12%

Streiger 2004

1-14

24

NT¶

-

-

-

-

C§ nR

4-24y 42%

(IHA,IIF,TAD) 0%

1-4a:75% 5-6a:64%

(xeno)

Page 36: proposal for the inclusion of benznidazol pediatric dosage

36

36

Reference Age (years)

Benznidazole Nifurtimox Design* Follow-up (months) Lost in FU (%)

Efficacy Measurements†

n Dose (mg/kg/d)

Time (days)

n Dose (mg/kg/d)

Time (days)

Serology Test (% neg)

Parasitology Test (% pos)

64 -

5 -

30 -

- 7

- 12-15

- 45-60

nB

} 31%

62% 86%

7-8a:58% 9-14a:43%

} 0%

Flores Chavez 2006

5-10

35

8

69

- - -

nC nR nB

12m -

(EIA) 6%

(xeno) 0%

(PCR) 14%

Duffy 2009

18d-18a

38

5-8

60

- - -

nC nR nB

18m Q-PCR a

30d & 60d

Not used (Q-PCR) 30d: 13%

60d: 5%

(PCR) 5%

Key

Design: C (controlled: control or comparison group), nC (not controlled); R (randomized), nR (not randomized); B (blind), nB (not blind)

† Efficacy Measurements: CFT (Complement Fixation Test), IHA (Indirect Hemagglutination, IIF (Indirect Immunofluorescence), EIA (Enzyme Immunoassay), Xeno (Xenodiagnosis), PCR (Polymerase Chain Reaction molecular assay), Q-PCR (Quantitative Polymerase Chain Reaction), atEIA (EIA trypomastigote antigen), F29EIA (EIA flagellate protein [F29]), TAD (Direct Agglutination Test)

¶ P: Placebo, NT: Not treated

‡ Analysis by Protocol (or Intention-to-treat without sensitivity analysis for the missing data)

§Selection bias in the experimental group (lower parasitemia than in the control group) Source: IRD 2009

Page 37: proposal for the inclusion of benznidazol pediatric dosage

37

37

Further to the data presented so far, there are data presented on children and adolescents

treated in the indeterminate chronic phase in field conditions in projects from the Doctors

Without Borders / Médécins Sans Frontières (MSF) reported by Yun et al.68 (2009). For 10

years, MSF implemented Chagas disease control programs for Chagas disease at Yoro,

Honduras (1999- 2002), Olapa, Guatemala (2003-2006), Entre Ríos, Bolivia (2002 – 2006) and

Sucre, Bolivia (2005-2008), focused on the diagnosis and treatment of patients up to 18

years old.

Diagnoses were confirmed by positive results in two different serology tests (conventional

ELISA and recombinant IHA, and IIF for indeterminate or discordant findings). Patients

testing positive for T. cruzi were treated with benznidazole 7.5 mg/kg/day administered two

or three times a day for 60 days. The efficacy of the treatment was assessed between 18

and 36 months post-treatment through convention ELISA serology, confirming the negative

findings through recombinant ELISA. According to the WHO protocol, patients presenting

two non-reactive serology tests (conventional and recombinant ELISA) on the same sample

and on the same day were rated as cured.

Table 12 summarizes some characteristics and findings obtained in each Program.

Table 12- Characteristics and findings obtained by the Chagas Disease Control Programs implemented by the MSF between 1999 and 2008.

Yoro, Honduras

Olapa, Guatemala

Entre Rios, Bolivia

Sucre, Bolivia

Program Duration

1999- 2002 2003-2006 2002 – 2006 2005-2008

Age range

<12 years < 15 years < 15 years < 18 years

Patients with confirmed diagnoses

232 124 1,475 1,145

Patients treated

232* 124 1,409 1,040

Negative seroconversion rate

87.1 58.1 5.4 0

* one patient with acute Chagas disease Source: Yun et al. (2009) There was a marked difference in results among the various programs. At Yoro, the

seroconversion rate was 87.1% (202/232) after 18 months of follow-up and 92.7% (215/232)

after 36 months, indicating that the treatment was extremely efficacious. In a more detailed

analysis of the Program at Yoro, Honduras, Escriba et al.69 (2009) showed an overall

seroconversion rate at 18 months of 88.2% (95% CI, 84-92.4%), rising to 93.9% (95% CI, 90.8-

97%) at three years. However, this increase was not statistically significantly. Of the 229

patients who were monitored for more than 18 months, 85.2% (95% CI, 80.5-89.8%)

presented a reaction in the antibody titrations (recombinant ELISA by optical density) of ≥

Page 38: proposal for the inclusion of benznidazol pediatric dosage

38

38

75% in relation to the initial values, and 93.4% (95% CI, 90.2-96.7%) after three years. This

difference is statistically significant. No material differences were noted by gender or age

range in the seroconversion rate and anti T. cruzi antibody titers.

At Olopa, the seroconversion rate after 18 months was 58.1% of patients for whom data

were available (25.5% of the cohort patients or 18/31), also suggesting that the treatment

was efficacious.

The seroconversion rates were lower at the two centers in Bolivia. In Entre Ríos, preliminary

findings indicated a seroconversion rate of 5.4% (59/1,101) post-treatment up to 60 months

post-treatment, with 950 patients monitored for a period of more than 18 months. The

seroconversion rates during the follow-up at 18 to 60 months were higher in the lower age

group (24.2% [16/66] among children < 5 years, 4.6% [14/305] in the age group of 5 to 9

years old and 1.9% [12/638] in the 10 to 14 years old age range). At Sucre, none of the 276

patients monitored for a period of 9 to 27 months post-treatment, presented

seroconversion.

According to Yun et al., these differences in the seroconversion rates may be explained by i)

the delay in negativisation by conventional serology, which may take between 5 to 10 years

in Latin America; ii) different susceptibility to treatment of the various parasite lineages

(predominance of type I T. cruzi in Central America type II in South America); iii) potential

differences in treatment efficacy as a function of the proximity of the acute phase; and iv)

constraints of data analyses due to in age groups and post-treatment follow-up times.

10.1.4 Population Pharmacokinetics Study in Children 0-12 years

There is an absolute lack of information on Benznidazole PK data for pediatric population

and its relationship with treatment safety and efficacy. In order to respond to this need,

DNDi and partners joined efforts in the design and implementation of this clinical trial

“Population Pharmacokinetics Study of Benznidazole in Children with Chagas’Disease”

(clinicaltrial.gov registry # NCT01549236; Appendix 8 – clinical study protocol).

The Principal Investigator of the study is Dr. Jaime Altcheh, a well-recognized specialist in pediatric Chagas disease. Five participating sites took part in this clinical trial (Hospital de Niños Ricardo Gutiérrez, Instituto Nacional de Parasitología in Buenos Aires, and sites in Jujuy, Salta and Santiago del Estero). The target study population of 80 patients was reached on 14-Jun-12 (LPI) and last patient last visit was performed on 10-Aug-12. This is the first pharmacokinetics study with benznidazole carried out in peadiatric population younger than 2 years old. The trial is designed as open, uncontrolled, single group assignment clinical trial, stratified by age groups of population pharmacokinetics study in children 1 day to 12 years old adopting treatments with Bz 12.5mg or 100mg Bz tablet (LAFEPE), 7.5 mg/Kg/day per oral in two daily doses, for 60 days.

Page 39: proposal for the inclusion of benznidazol pediatric dosage

39

39

Newborns-2 years-old children have been included as they represent the population of

congenital cases. Children 2-12 years-old have also been included as the target population,

to represent those who may have been infected via congenital or vector-borne transmission,

and usually present with the early chronic indeterminate form of the disease.

Population pharmacokinetics has been chosen as the study design as it would minimise the

number of samples in the pediatric population. The dearth of PK in adults and lack of

information on the variability in the target population does not allow for power calculations

and the use of optimal sampling design for definition of the timing of samples. Experts

reviewed the available information and recommended sparse sampling, with 4 PK samples

distributed over the absorption phase (1 sample), steady-state (2 samples) and elimination

phase (1 sample), and 2 additional micro-samples collected in filter-paper at steady-state.

With a total of 6 PK measurements per patient and a total of 80 patients stratified by age, it

is expected that PK curves and variability can be drawn with an adequate level of precision.

Diagnosis of T. cruzi infection was confirmed at entry by direct microscopic examination or at least two positive conventional serologies (ELISA, IIF or HAI). Subject enrolment was stratified by age GROUPS: 41 patients in the group of newborns to 2 years (minimum of 8 newborns) and 40 patients in the group of > 2-12 years. For the pharmacokinetics (PK) sampling five blood samples per patient, representing a total of approximately 100µL of blood, were collected in filter-paper at randomly pre-assigned time-points as follows: • Day 0 (absorption phase one sample at randomly selected time-point 2-5hs after first dose), • Steady State phase (two samples to be collected: one sample at Day 7, and another at day 30; both at randomly selected time-points from pre-dose to 8 hours post-dose) • End of treatment AT DAY 60 (two samples at randomly selected time-points 12 – 24 hours after last dose. Benznidazole in plasma was measured by HPLC-MS-MS and POP-PK modeling was performed with NONMEM software (nonlinear mixed effects analysis). Clinical study report is currently in preparation. A total of 83 patients were screened for the

study, of which 2 patients resulted in screening failures and a total of 81 subjects enrolled.

Seventy six (76) patients completed the study treatment and 5 subjects discontinued. PCR

analysis showed no treatment failures, with 100% negativisation at end of treatment.

10.1.5 Conclusions on efficacy

Despite the heterogeneity of the studies presented in terms of objectives, geographic

location, age ranges, numbers of children included in these studies, therapeutic schemes

used, duration of post-treatment monitoring and the cure control tests deployed, there is

clear evidence of the efficacy of benznidazole for treatment of children infected by T. cruzi,

Page 40: proposal for the inclusion of benznidazol pediatric dosage

40

40

particularly those less than a year old, targeted by the pediatric formulation, and suggesting

greater efficacy for early treatment.

For the treatment of congenital infections, the serum-negative rates vary from 87%

(Schijman et al., 2003) at 36 months (100% in the 0 to 3 months old group) to 100%

(Russomando et al., 1998) at 24 months in children up to two years old.

For the treatment of acute infections, these rates vary from 76% (Cançado et al., 2002) at 13

years in children under 10 years old to 100% (Ferreira, 1988) at 15 years in children between

2 and 18 years old.

For early chronic phase treatment, the studies show significant variability, with a serological

cure rate than may be less than 10% (Yun et al., 2009; Ferreira, 1990; Flores-Chavez et al.,

2006) but possibly over 60%. At this phase of the disease, it is difficult to determine

serological response, as conventional serology may remain positive for a period of 5 to 10

years after treatment. Two placebo-controlled randomized clinical trials conducted with

children between 6 and 12 years old, with follow-up periods lasting several years, presented

very positive outcomes, with seroconversion rates of around 60% (Sosa Estani et al., 1998;

follow-up at 48 months; de Andrade et. Al, 1996; follow-up at 36 months), reaching 89% at 6

years old (de Andrade et al, 2004) and 77% at 9 years old (Sosa Estani et al., 2002).

Finally, as it is described in section 11.7 (Update information about safety preliminary results

of Population-Pharmacokinetics study in Argentina) the study just concluded in Argentina

which brings evidence in relation to efficacy of benznidazole 12.5mg in children younger

than 2 years old (including newborns) showing 100% cure rates measured by PCR after

treatment.

11. Summary of comparative evidence on safety*:

11.1 Safety Overview

11.1.1 Introduction and Overview

The literature review presented in this section was presented as part of the dossier for

submission for marketing authorization (registration) by the Brazilian Drug Regulatory

Authority (ANVISA) in March 2011. Additional information and recent publications were

incorporated in this summary.

Considering that benznidazole is a medication with well-established use for the treatment of

Chagas disease, with well documented efficacy, safety and tolerability in the literature, and

with the adult tablet already registered by the Brazil's Drug Regulatory Authority (ANVISA).

In addition, as the pediatric formulation consists of the same qualitative composition as the

adult 100 mg tablet, and as its dosage follows the recommendations followed by the

Brazilian Ministry of Health and the WHO (5 – 10 mg/kg/day administered twice a day for 60

Page 41: proposal for the inclusion of benznidazol pediatric dosage

41

41

days), in vivo clinical trials were not deemed necessary to support the registration of the new

pediatric dosage form.

The dossier confirming the clinical safety of the new pediatric tablet consists of a review of

published benznidazole safety data in general, with a comparison of the individual data

resulting from clinical trials resulting from clinical trials conducted with the adult population

and the pediatric population that is the target, indicating better tolerability for this

medication in the pediatric population.

However, as mentioned previously, the pediatric formulation development plan includes

conducting in vivo clinical pharmacology trials, during which aspects related to clinical

tolerance and safety were observed and analyzed, in order to contribute to the evidence

already available in the literature on the pediatric population, monitoring the frequency and

intensity of adverse reactions in children and / or notifying new adverse events. A safety and

tolerability assessment plan under usage conditions will also be implemented, as part of the

risk management plan presented to ANVISA.

11.2 Safety and Tolerability of Benznidazole

The tolerability of benznidazole is considered as generally better in children and for patients

in the acute phase, regardless of age range, than for adults in the chronic phase. There is

limited evidence regarding the safety of benznidazole for children, particularly with

congenital and acute infections. However, the reported data indicates excellent tolerability

in children under one year old, with no significant of adverse reactions.

Most of the existing data relates to tolerance and safety in children during the indeterminate

chronic phase and for adults in the chronic phase. For children in the indeterminate phase,

good tolerability has been demonstrated, with treatment interruption rates due to adverse

events varying from 0% (Steiger et al.Error! Bookmark not defined., 200470; Sosa Estani et

al.Error! Bookmark not defined., 2002 ) to 10% (Sosa Estani et al.,Error! Bookmark not

defined. 1998), while for adults in the chronic phase, the treatment drop-out rate due to the

appearance of adverse reactions is generally higher, although varying by author: 0%

(Sanchez et al.,71 2008; Viotti et al.,72 1994), 9.5% (Pinazo et al.,73 2010), 11.5 % (Coura et al.,

1997)74, 13% (Viotti et al., 2006)75, 18% (Gallerano et al., 200076; Fabbro et al., 2007; Sosa

Estani et al.,77 2004), 25% (de Pontes et al.,78 2010) and 41 % (Levi et al.79 1996), indicating

greater intolerance for benznidazole.

11.3 Adverse effects during treatment with Benznidazole

Adverse reactions are frequent, generally occurring in 25% to 30% of patients80. Due to

toxicity, treatment is not recommended during pregnancy, and for women of child-bearing

Page 42: proposal for the inclusion of benznidazol pediatric dosage

42

42

age not taking contraceptives, except for severe acute cases. Indications for patients with

severe disease must be carefully assesed83,81.

The most frequent adverse reactions are dermopathies due to hypersensitivity that are not

dose-dependent, generally appearing on the ninth day of treatment, and expressed in the

form of pruriginous, non-blistering polymorphous erythema, followed by scaling and peeling.

In < 20% of patients, the dermopathies are mild, not requiring the interruption of the

treatment. In < 5%, they may be moderate, accompanies by fever and purpura, with the

recommended temporary interruption of treatment, to be reintroduced as indicated by

clinical tolerance after treatment with antihistamines or corticosteroids. In < 1% of patients,

dermopathies may be severe, requiring the suspension of the treatment.

Another frequent adverse reaction is dose-dependent peripheral polyneuropathy, generally

appearing after the fifth week of treatment. This occurs in less than 1% of cases, appearing

in the form of paresthesia or peripheral polyneuritis symptoms, particularly after prolonged

treatment. It is advisable to interrupt the treatment until the symptoms improve.

Gastrointestinal disorders, such as nausea, vomiting, diarrhea and intestinal cramps, may

occur during the initial stage of treatment in around 10% of cases, and must receive

symptomatic clinical treatment, with no need to interrupt the treatment of lower the dose.

Other side effects are rare, such as ageusia, which may occur in less than 0.5% of cases,

generally towards the end of treatment, and medullar hypoplasia, which occurs in <1% of

cases, generally between the 20th and 30th day of treatment. In case of ageusia or

leukopenia, granulocytopenia, neutropenia, agranulocytosis and thrombocytopenia, the

treatment must be interrupted.

Some authors also noted other adverse reactions such as headache, dizziness, fatigue,

arthralgias, generalized edema or in the extremities, anorexia, and an increase in hepatic

enzymes. Table 13 summarizes the adverse events identified in the literature.

Table 13- Main adverse events observed during treatment with benznidazole *

Systems and Organs

Symptoms

Dermatological

Maculo-papular cutaneous eruptions

Erythematous Plaques

Rash

Itching

Blistering eruptions

Peeling skin

Neurological (central and peripheral nervous system)

Paresthesia

Hypoesthesia

Tremors

Dizziness

Headaches

Psychiatric manifestations

Gastro-intestinal Nausea

Page 43: proposal for the inclusion of benznidazol pediatric dosage

43

43

Vomiting

Diarrhea

Abdominal pain

Epigastric pain

Anorexia

Dry mouth

Ageusia

Skeletal Muscle

Arthralgia

Myalgia

General / Constitutional Symptoms

Fever

Asthenia

Increased appetite

Lymphatic Generalized edema

Edema in the extremities

Lymphadenopathy

Bone Marrow

Leukopenia

Thrombocytopenia

Granulocytopenia,

Neutropenia,

Agranulocytosis

Metabolism / laboratory Alteration to the hepatic enzymes

* based on classification defined by the National Cancer Institute’s Common Terminology Criteria for Adverse Events (CTCAE version 3.0).

One author also reported a case of jaundice with alterations to the bilirubin dosing in the

serum after preventive treatment with benznidazole of a laboratory accident.82

Another author reported a case of disabling pain syndrome in a Bolivian woman (31 years

old) in the course of treatment with benznidazole.83

The teratogenetic potential of benznidazole has been reported in some in vivo trials with

animal models, confirming that treatment is counter-indicated for pregnant women and

women of child-bearing age not taking some type of contraceptive.84,85,86

The toxicity of benznidazole in several organs has also been described in vivo with animal

models. 87,87,88,89,90

With regard to the potential genotoxicity of do benznidazole, some authors report the

induction of chromosome aberrations during in vivo trials with animal models85,91,92,93 and in

human cells in vitro, 87,94,95 at doses close to the plasma concentration limits of treated

patients.97 Gorla et al. (1988) noted a slight but significant increase in micronucleus

formation and the appearance of chromosome aberrations in lymphocyte cultures for two

groups of children with Chagas disease, before and after treatment with benznidazole.87,98

At the same time, Gorla & Castro (1985) did not report any significant increase in the

formation of micronuclei in the bone marrow or spleen lymphocytes of rats treated with

benznidazole, 87,96 while Souza et al. (1991) did not report any significant increase in the

Page 44: proposal for the inclusion of benznidazol pediatric dosage

44

44

frequency of chromosome aberrations in bone marrow cells of the appearance of

micronuclei in the peripheral blood cells of rodents receiving different dosages and with

different exposure times to benznidazole.97 Similarly, the clastogenic and mutagenic effects

were reversible, and were not accompanies by any clinical manifestations in the study

conducted by Moyá & Trombotto (1988) of children treated with benznidazole and

nifurtimox98.

Other authors also identified an increased risk of developing lymphomas in animal models in

vivo.99,100 However, no increase in the rate of human lymphomas was reported among the

large number of patients treated, despite a higher rate of neoplasias reported in the few

patients infected by T. cruzi with heart transplants.101

The toxicity of benznidazole is closely linked to its trypanocide mechanism, deployed

through a covalent binding of free radicals produced by the nitro-reduction of macro-

molecules and their interaction with the host DNA. 87, 96

11.4 Adverse events in adults

Adverse events occur frequently among adults taking benznidazole. However, among adults

treated with benznidazole, the frequency and type of adverse events most frequently

notified varied among authors.

In the study conducted by Gallerano et al. (2000) of 130 patients between 10 and 79 years

old (average age 33, 4 + 14.2) treated with benznidazole at 4 to 8 mg/kg/day, for 45 to 60

days, adverse events were observed in 32% of patients treated with benznidazole, mainly

gastric intolerance, cutaneous eruptions and peripheral neuropathy, also observed in the

study by Coura et al. (1997), including 26 patients receiving benznidazole at doses of

5m/kg/day for 30 days. However, in this latter study, the authors identified the appearance

of mild adverse events in 29.1% patients treated with placebo (aerophagia, post-prandial

fullness, anorexia, headache, drowsiness and lassitude), with two patients (8.3%)

abandoning treatment.

Fabbro et al. (2007) reported adverse events in 27% of 33 patients treated with

benznidazole at a dose of 5m/kg/day for 30 days, consisting of maculo-papular erythema

(18% or 6/33), edema (9% or 3/33), nausea (3% or 1/33), headache (3% or 1/33), itching (3%

or 1/33) and a slight increase in the hepatic transaminases (3% or 1/33).

The main adverse effects noted by Viotti et al. (2006) in 283 patients between 30 and 50

years old completing treatment with benznidazole at 5m/kg/day for 30 days (55/246 or 22%)

were allergic dermatopathy; mild (36 patients or 14.6%), and moderate (2 patients or 0.8%),

gastrointestinal disorders (11 patients or 4.5%); headache (3 patients or 1.2%); itching (2

patients or 0.8%) and fever (1 patient or 0.4%). Also in this study, 33 of the 37 patients who

interrupted the treatment, dropped out due to the appearance of severe allergic

dermatopathy, while the other 4 were prompted by gastrointestinal disorders.

Page 45: proposal for the inclusion of benznidazol pediatric dosage

45

45

In another study by Viotti et al. (1994) of 131 patients, with an average age of 46 years,

treated with benznidazole (5 mg/kg/day) during 30 days, adverse reactions were noted in

20% of those patients, with the most frequent being moderate allergic dermatitis (77%),

gastrointestinal intolerance (16%), widespread allergic dermatitis (7%). Other less significant

effects were headache, itching and edema of the lower extremities..

The frequency of the adverse reactions noted in these studies matches the estimated

frequency in the Brazilian Consensus on Chagas Disease, hovering between 20% and 30%.

However, three studies identified higher rates of adverse reactions in adults.

In the study conducted by Pinazo et al. (2010) of 105 patients between 16 and 58 years old

(average age 38.7 years) treated with benznidazole at 5 mg/kg/day for 60 days, 57.1%

(60/105) presented adverse reactions, with 47% of them (27/60) presenting more than one

adverse reaction. The most frequent adverse reactions were headache (56.2% or 59/105

patients), dermatopathies (50.5% or 53/105 patients), notably urticaria, rash and itching,

anorexia (40% or 42/105 patients), joint disorders (36.2% or 38/105 patients), asthenia (30%

or 32/105 patients), paresthesias (27.6% or 29/105 patients) and gastrointestinal disorders

(15% or 16/105 patients), mainly epigastralgia. Ten patients halted the treatment due to the

appearance of adverse reactions, eight of whom presented severe urticaria and fever, with

two dropping out due to digestive intolerance.

In a study of 18 patients between 19 and 41 years old (average age 25 years) treated with

benznidazole at 5mg/kg/day for 60 days, Sanchez et al. (2008) noted adverse events in 67%

of these patients, consisting of dermatopathy due to hypersensitivity (39%), headache

(33%), gastrointestinal disorders (22%), arthralgia and peripheral neuropathy (11%), with a

slight increase in the hepatic enzymes (11%) and asthenia (6%).

In a study conducted by Pontes et al. (2010) with evaluation of adverse reactions in 32

adults patients treated with benznidazole at 5 mg/kg/day, for 60 days, adverse events were

observed in 87.5% of these patients. This study identified 20 different types of adverse

reactions, with the most frequent being itching (50%), paresthesia (43.8%), asthenia (37.5%),

headache (34.4%) cutaneous rash (31.3%) and peeling skin (25%). The dermatological

system was the most severely affected, with 35% of the symptoms, followed by the central

and peripheral nervous systems with 22% of the reported symptoms.

Differences in the frequency in the adverse reactions rates among these studies may be due

to the prolonged durations of the treatment in the last three of those trials mentioned

above (60 days versus 30 days).

11.5 Adverse events in children

As mentioned previously, the data available in the literature indicated better tolerability for

benznidazole among children, with fewer adverse effects appearing, compared to adults.

The evidence is detailed below taken from studies conducted with children with congenital

infections, during the acute and early chronic phases of the disease.

Page 46: proposal for the inclusion of benznidazol pediatric dosage

46

46

11.5.1 Children with congenital infections

In the study conducted in Bolivia by the IRD of 111 new-born infants diagnosed with

congenital infections at birth and treated with benznidazole (59 with a therapeutic scheme

of 2.5 mg⁄ kg twice a day for 60 days and 52 with therapeutic scheme of 7.5 mg⁄ kg once a

day for 30 days), no reports were found of any adverse reactions (Chippaux, et al., 2010). A

similar finding was reported in the study by Russomando et al. (1998) that did not identify

any adverse reaction or toxicity when monitoring six children treated with benznidazole (7

mg/kg and per day, taken twice a day for 60 days).

Other studies of children with congenital infections addressed in the efficacy analysis section

did not provide sufficient information on the safety and tolerability aspects of benznidazole.

Despite the limited amount of information, these studies indicate the excellent tolerability

and safety of benznidazole in new-born infants with congenital infections.

The authors suggest that the nitro-reduction metabolism capacity of new-borns, which is

responsible for toxicity, is low, while susceptibility to T.cruzi remains unaltered.87,101,102. This

would explain the excellent tolerability of benznidazole among babies, despite the excessive

high doses to which they are exposed, due to inaccurate splitting of the 100 mg tablet.

11.5.2 Children with acute infections

In the study conducted by Barclay et al. (1978) of 139 children (ages not stated) with acute

infections treated with two therapeutic schemes (benznidazole, progressive doses of up to

7.5 mg-10 mg/ kg/day for 30 days and benznidazole at a uniform dose of 5 mg/kg/ day for 30

days), adverse events were noted in 18% of these children, with 23 of them presenting

dermatological manifestations (morbilliform exanthema), none of them severe, and

vanishing with the temporary suspension of the treatment for a few days; and two children

with de complaints of arthralgia, although with no signs of inflammation.Error! Bookmark

not defined.

Other studies in children with acute infections addressed in the efficacy analysis section did

not mention the safety and tolerability aspects of benznidazole related directly to the study.

11.5.3 Children with indeterminate chronic infections

The literature provides more information on adverse reactions occurring in children at the

early indeterminate phase. We highlight and detail six studies below.

In a double-blind randomized, control clinical trial conducted by Andrade et al. (1996) of 129

children between 7 and 12 years old, 64 were treated with benznidazole at 7.5 mg/kg/day

for 60 days, and 65 with placebo. The treatment was well tolerated, interrupted for only

one patient (1.6%) due to the appearance of maculo-papular cutaneous eruptions and

severe itching. Dermatological reactions occurred more frequently in the experimental

Page 47: proposal for the inclusion of benznidazol pediatric dosage

47

47

group than in the control group (8 [12.5%] vs 2 respectively p= 0.05). Minor adverse events

such as nausea, anorexia, headache, epigastric pain and arthralgia occurred in less than 5%

of these patients, with no significant differences between the two groups. No signs of

toxicity were identified. The anemia rate (Hb < 110g/L) proved similar in both groups, and

no patient developed leukopenia or neutropenia. Hepatic and renal function tests remained

within the normal limits throughout the entire study.

During the double-blind randomized control-case clinical trial conducted by Sosa Estani et al.

(1998) of 106 children between 6 and 12 years old, with 55 of them treated with

benznidazole at 5 mg/kg/day for 60 days and 51 children with placebo, the treatment was

well tolerated, although it had to be interrupted due to moderate adverse events for six

patients in the experimental group (10%). The symptoms regressed after suspending the

treatment. Less than 20% of these patients reported adverse events that included intestinal

cramps, maculo-papular cutaneous eruption, headache, anorexia, vomiting, nausea,

diarrhea, dizziness, paresthesia and mild hand tremors. However, a significant difference

between the two groups was identified only with regard to intestinal cramps and cutaneous

eruptions (p=0.05) which appeared respectively at around the 11th and 19th days of

treatment. No severe adverse events were notified for any patient. Additionally, no signs of

toxicity were observed, with the laboratory examinations remaining within the normal limits

for both groups.

In a supplementary follow-up study of the serological progress of 252 Argentine children

treated with benznidazole, including 46 of the children participating in the clinical trial

mentioned above, Sosa Estani et al. (2002) did not identify any long-term adverse events

that could be attributed to benznidazole in this group. In a group of N 147 children between

1 and 14 years old, treated for 30 days in 1994, 4.8% of the children presented some adverse

reaction during treatment, and in the group of 40 children between 1 and 14 years old

treated for 30 days in 1995, no adverse events were notified. In both these groups,

treatment was not interrupted due to adverse reactions.

In a cohort study conducted with 95 children between 1 and 14 years old, with 64 of them

treated with benznidazole at 5 mg/kg/day, split into two daily doss for 30 days, Streiger et al.

(2004) noted good tolerability for the treatment which had to be interrupted for two

children (2/53 or 3.8% of children who had at least one post-treatment control) due to

benznidazole intolerance. In the group treated with benznidazole, the authors identified

adverse events such as vomiting, widespread erythema with edema and itching without

specifying the level of severity nor the frequency of occurrence.

In the course of the Chagas Disease Control Program implemented by the MSF at Yoro,

Honduras, Olapa, Guatemala, Entre Rios and Sucre, Bolivia, focused on the diagnosis and

treatment of patients up to 18 years, adverse reactions to benznidazole were recorded by

type and severity.

Table 14 presents a detailed listing of the number of patients with adverse events by

program and severity level.

Page 48: proposal for the inclusion of benznidazol pediatric dosage

48

48

Table 14 – Adverse events by program and severity level - MSF

Program Adverse events

Nº of patients

% x total patients treated

Nº of patients interrupting treatment due to adverse events (%)

Mild %

Moderate %

Severe %

Yoro Honduras

116 50.2 3/231 (1.3%)

97.4 0 2.6

Olopa Guatemala

63 50.8 80.8 14.3 4.8

Entre Rıos Bolivia

361 25.6 28/1409 (2%)

79.5 18.8 1.7

Sucre Bolivia

394 37.9 61/1040 (5.8%)

61.4 28.2 10.4

Source: Yun et al. (2009)

A higher rate of adverse reactions was noted in the programs conducted in Central America,

compared with the Bolivian programs. However, most of the adverse events were mild, with

a higher proportion of severe events (10.8%) noted in Sucre. The proportion of patients

interrupting treatment due to adverse events is low (<6%), indicating good tolerability for

benznidazole.

Severe reactions reported at Yoro were neurological (neuromuscular disorders of the lower

limbs appearing after six weeks of treatment in three patients); neuromuscular (2/63) and

dermatological (1/63) in Olopa. In Entre Rios and Sucre severe adverse reactions occurred in

6 and 41 patients respectively, particularly a case of Lyell’s syndrome in a 13-year old girl,

which occurred in the 34th day of treatment, and 1 case of Stevens Johnson syndrome.

At Yoro, the most frequent adverse reactions were 26 gastrointestinal disorders (.8%),

mainly epigastralgia and abdominal pain, and to a lesser extent nausea and / or vomiting and

anorexia, followed by cutaneous reactions (13%, mainly itching and maculo-papular

exanthema less frequently), as well as neurological (10.4%). At Olopa, the most frequent

adverse reactions were dermatological (26%), gastrointestinal (25%), neuromuscular (23%)

and assorted (26%). At Entre Rios, the main adverse reactions were dermatological (56%),

gastrointestinal (25%), neuromuscular (18%), of which 11% were mixed. At Sucre the

adverse events were predominantly dermatological (68.5%).

The authors also identified a significant difference in the proportion of adverse reactions by

age brackets (chi-square test) in the two Bolivian centers. This difference was not noted at

Yoro and Olapa.

In the Bolivian programs, the risk of developing adverse reactions to the treatment rose with

age (12% of adverse reactions in children under five years old, compared to 25% in the age

Page 49: proposal for the inclusion of benznidazol pediatric dosage

49

49

range between 10 and 14 years at Entre Rios; 13.4% in children under five years old,

compared to 50% in the age bracket between 15 and 18 years old at Sucre). And also at

Sucre, a higher treatment interruption rate was noted, due to adverse effects among older

children (0% in children under five years old, compared to 8.6% in the age range between 15

and 18 years old) (Yun et al., 2009).

These evidences corroborate the greater tolerability and safety of benznidazole among small

children, noted by several authors.

Another prospective cohort study published recently, conducted between 2003 and 2007 at

the Ricardo Gutierrez Pediatric Hospital in Buenos Aires (Argentina) by Altcheh et al. 2011

described adverse events in 107 children between 10 days and 19 years old (average of age

6.9 years), diagnosed with asymptomatic infections of T. cruzi, treated with benznidazole at

5 to 8 mg/kg/day, administered two or three times a day for 60 days, and monitored for a

period of three years. Among them, 62 adverse events were noted, related to treatment in

44 children (41.1% of patients), mostly mild (80.6%) and moderate (16%). Only two adverse

events (3.2%) were rated as severe (widespread rash). In this cohort, seven patients (6.5%)

abandoned the treatment due to the adverse events (6 dermatological and one

gastrointestinal), while six of them were more than seven years old. The adverse events

resulted in the temporary suspension of treatment for seven children (4 due to the

appearance of rash, 2 for gastrointestinal discomfort and 1 for headache), although all of

them later returned and completed the treatment with no other interruptions. Among

them, 71% of the adverse events noted were clinical, being dermatological for 21% of the

cases (22/107 children), mainly rash and eczema; gastrointestinal in 8.5% of these children

(9/107); neurological (central nervous system) in 9% of these children (10/107), mainly

headaches; and neuromuscular for 2.8% of these children (3/107). Alterations in the

laboratory examinations accounted for 29% of the adverse events recorded, with the most

frequent being hematological (12 occurrences), mainly eosinophilia and leukopenia, and

metabolic (6 occurrences), mainly an increase in hepatic enzymes.

The average duration of the adverse events was 8.2 days (95% CI: 4.1–12) and in 73% of the

patients, they occurred during the first ten days of treatment, with some differences in the

average appearance time due to the type of event. Neurological adverse events appeared

during the first two days, on average, followed by gastrointestinal events (average of five

days) and dermatological events (average of nine days, with a statistically significant

difference compared to the neurological events).

The statistical analysis also showed that the occurrence of adverse events varied by age,

being more frequent among older children. The average age of the children presenting

adverse reactions was 9.9 years (95% CI: 8.2–12), significantly higher than the average age of

the children with no adverse events (4.8 years [95% CI: 3.7– 6.0]; p<.001, t test). Moreover,

77.3% of the adverse events occurred in children more than seven years old, while only 18%

of children less than two years old (7/38) presented adverse events, at rates significantly

lower than those noted for older children (18% vs 53%; P <.001, test X2).103

Page 50: proposal for the inclusion of benznidazol pediatric dosage

50

50

This evidence matches the observation in the studies presented previously, in terms of the

better tolerability of benznidazole in children, with the occurrence of a higher proportion of

mild adverse events, and the increased risk of their appearance with rising age, with the risk

being lower in young children, who are the target public of the pediatric formulations.

Table 15 summarized the benznidazole safety and tolerability data for children in the studies

presented above.

Page 51: proposal for the inclusion of benznidazol pediatric dosage

51

51

Table 15 – Overview of the benznidazole safety and tolerability data for children

Reference Phase

Age (in years) Sample

Dose (mg/kg/d)

Time (days)

Administration/ day

Adherence (%)

Number (%) of e patients

abandoning treatment due to AE

Main adverse events and number

(%) of patients

Number of severe

adverse events (%)

Chippaux (IRD)

2008 -2009

Congenital (< one month)

59 52

5 7.5

60 30

2 1

80 (≥55d) 76 (≥25d)

? - -

0 0

Russomando 1998

Congenital (<2 y)

6 7-10 60 2 ?

0 - 0

Barclay 1978

Acute

Children (?)

107 30

3-10†

5

30 30

? ?

- -

? } dermatological

23/137 (17%) - -

de Andrade 1996

Chronic indeterminate

(7-12)

64

7.5

60

2 98 1/64 (1.6%)

Dermatological 8/64 (12.5%)

Digestive and assorted (5%)

1 (1.6%)

Sosa Estani 1998

Chronic indeterminate

(6-12)

55

5

60

? 90 6/55 (10%)

Dermatological Digestive (20%?)

0

Sosa Estani 2002

Chronic indeterminate

(1-14)

137 40

5 5

30 30

? ?

0 0

4.8% -

0 0

Page 52: proposal for the inclusion of benznidazol pediatric dosage

52

52

Reference Phase

Age (in years) Sample

Dose (mg/kg/d)

Time (days)

Administration/ day

Adherence (%)

Number (%) of e patients

abandoning treatment due to AE

Main adverse events and number

(%) of patients

Number of severe

adverse events (%)

Streiger 2004

Chronic indeterminate

(1 - 14) 53 5 30 2 87

2/53 (3.8%)

Dermatological

Digestive

2 (3.8%)

Yun 2009 (MSF)

Chronic (Yoro <12 y)

231

}7,5

}60

}2-3

98.7

3/231 (1.3%)

116/231 (50.2%) Digestive (26.8%) Dermatological

(13%) Neuromuscular

(10.4%)

3 (1.3%) (2.6%)*

neuromuscular

Chronic (Olopa <15 y)

123

95

?

63/123 (50.8%) Dermatological

(26%) Digestive (25%) Neuromuscular

(23%) Assorted (26%)

3 (2.3%) (4.8%)*

neuromuscular (2)

dermatological (1)

Chronic (E. Rios <

15y)

1,409 90.5 28/1409

(2%)

361/1.409 (25.6%) <5a:12%

10-14y:25% Dermatological

(56%) Digestive (25%), Neuromuscular

6 (0.4%) (1.7%)*

Page 53: proposal for the inclusion of benznidazol pediatric dosage

53

53

Reference Phase

Age (in years) Sample

Dose (mg/kg/d)

Time (days)

Administration/ day

Adherence (%)

Number (%) of e patients

abandoning treatment due to AE

Main adverse events and number

(%) of patients

Number of severe

adverse events (%)

(adverse events18%),

Chronic (Sucre <18y)

1,040 87.7

61/1040 (5.8%) <5a: 0%

15-18a:8.6%

394/1.040 (37.9%) 5a:13.4%

105-18y:50% Dermatological

(68.5%)

41 (3.9%) (10.4%)*

Altcheh, 2011

Asymptomatic (10 d – 19 y)

107 5-8 60 2-3 85.0

7/107 (6.5%) < 7 a: 1 (0.9%) >7ª: 6 (5.6%)

44/107 (41.1%) < 2a: 18% >2ª:53%

Dermatological (21%)

Digestive (8.5%) Neurological (CNS)

(9%) Neuromuscular

(2.8%) Laboratory (29% of

AE)

2 (3.2%) dermatological

Key Adherence = Percentage of treatments completed † daily dose increased from 3 ­to 7.5-10 mg/kg/day during the first 12 days and maintained at 7.5-10 mg/kg/day for a further 18 days * Percentage x total adverse events

Page 54: proposal for the inclusion of benznidazol pediatric dosage

54

11.6 Conclusions on Safety and Tolerability

Despite the countless adverse reactions reported in the literature prompted by

treatment with benznidazole, this is one of the only two treatment options available

for Chagas disease, and is generally better tolerated than nifurtimox.

The data presented in this section shows better tolerability for benznidazole among

children than for adults, with treatment abandonment rates due to adverse events

being rare among new-borns, and no more than a maximum of 10% in children at the

indeterminate phase, while these rates may rise to 40% among adults, usually hovering

at around 20%.

Similarly, differences are noted in the frequency with which adverse reactions appear,

comparing children and adults. While for the latter adverse reactions occur on

average in between 20% and 30% of the patients, and may reach >60% with long-

duration therapeutic schemes, the studies did not note any adverse reactions in new-

borns. Among children of the indeterminate phase, the frequency with which adverse

reactions appear varies between 5% and 50%, depending on the study, remaining

within the standards noted in adults, although the scope of the age brackets

encompassed by the studies is heterogeneous. However, the authors analyzing the

occurrence of adverse reactions by age bracket (Yun et al., 2009; Altcheh et al.,

manuscript accepted for publication), noted significant differences, with lower

occurrence rates among young children, indicating high risks of adverse reactions at

older ages.

Furthermore, fewer neurological events were noted among children, who presented

mainly dermatological and gastrointestinal adverse events, in contrast to adults.

It is thus believes that the pediatric formulation of benznidazole will offer good

tolerability and safety, as its target public is less likely to present adverse events, while

also allowing more accurate dosing within the established therapeutic margins of 5-10

mg/kg/day, in addition to reducing the risk of dose-dependent adverse events.

11.7 Update information about safety – preliminary results of POP-PK study in

Argentina

As presented in Section 10.1.4, the “Population Pharmacokinetics Study of Benznidazole in

Children with Chagas’Disease” started in April 2011 in Argentina as part of the follow-up

strategy for the development of the pediatric formulation benznidazole 12,5mg. Details of

study design were described earlier.

Page 55: proposal for the inclusion of benznidazol pediatric dosage

55

Preliminary results were presented in September 2012 at the International Congress of

Tropical Medicine and Malaria. A total of 83 patients were screened for the study, of which 2

patients resulted in screening failures and a total of 81 subjects enrolled. seventy six (76)

patients completed the study treatment and 5 subjects discontinued.

Serious Adverse Event (SAE)’s occurred in 3 children (01 bronchitis, 01 gastroenteritis, 01

maculopapular exanthema) and the only the latter event was considered possibly related to

the study drug. This subject was withdrawn from the study and the rash discontinued with

treatment interruption. Adverse drug reactions were identified in 30 cases (14 rash, 11

gastroenteritis, 4 lab abnormalities and 1 maculopapular exanthema) and all recovered well.

The age distribution of adverse events parallels previously published studies with lower

occurrence rates among the very young, indicating high risks of adverse reactions at the older

age group (2-12 years of age).

From those 76 patients who completed treatment, 45 children received Bz 12,5mg and 386

samples were collected for PK analysis. Close out visits have already taken place for all five

participating sites and final study report is in preparation.

12. Summary of available data on comparative cost** and cost-effectiveness within the pharmacological class or therapeutic group:

As presented in previous questions of this questionnaire, the two medicines indicated

for the etiological treatment of Chagas disease are benznidazole and nifurtimox.

According to the recent published WHO recommendations (October, 2011) for

congenital Chagas disease, both medicines are indicated for the treatment of those

cases.

Nifurtimox is provided by Bayer to WHO through a donation agreementciv. WHO

provides the medicines to countries free of charge.

Benznidazol tablet 12.5mg is a single-source medicine produced by Lafepe.

The development of this dosage form was a result of a partnership with DNDi

established in 2008, leading to:

The product available as package with 24 blisters of 10 tablets per blister, with

a total of 240 tablets per package. This amount can be considered as one

treatment for a child.

A cost of US$ 7 per package of 240 tablets, excluding transportation cost.

The reference price available for the adult dosage form produced by Lafepe was made

public (July, 2012) at Brazilian Reais (R$) of 0,338 per tablet, which roughly means US$

16,692/100 tablets (the adult package is 10 blisters of 10 tablets/blister).

2 Considering in 2012 currency rate US$1=R$2.

Page 56: proposal for the inclusion of benznidazol pediatric dosage

56

In relation to cost-effectiveness evidence, two publications were identified in the

literature. They do not focus to the comparison of the two existing medicines

(benznidazole and nifurtimox) for congenital Chagas disease cases, but rather on

adoption of screening programs for congenital Chagas diseasecv,cvi.

Billot et al. (2005) studied the cost effectiveness of a control program of congenital

Chagas disease in Bolivia. It was demonstrated that US$ 21 million per year for 2,718

infected new-borns reflected the direct and indirect costs estimated from the disease

complications and death, from birth to adulthood. If a control program would detect

and treat all cases, this would mean an investment of US$ 123 per new-born with the

infection or US$ 1.2 per new-born.

Sicuri et al. (2011) developed an economic evaluation of to compare the adoption or

not of an Active Detection of the Infection (ADI) of all pregnant women, as well as

new-borns, from Latin America countries who are living in Spain. Several variables

were adopted for this comparison and results were measured considering also QALYs

(Quality-Adjusted Life Year) gained. Benznidazole was the medicine used for the cases

of treatment. Comparison was made in relation to “test” or “not test” the mothers and

also in relation to the newborn. Results have shown that it is more cost-effective to

screen both mothers and newborns. For the first comparison – test or not test the

mothers - the cost effectiveness ratio was 96 D /QALYs gained in the case of screening

and 1675 D /QALYs gained in the case of no screening. For the second comparison,

cost effectiveness was 22 D /QALYs gained in the case of screening the newborns and

125 D /QALYs gained in the case of no screening.

13. Summary of regulatory status of the medicine (in country of origin, and preferably in other countries as well)

Benznidazole 12.5mg was registered for the treatment of infections due to

Trypanosoma cruzi. The product was approved as a new dosage form 12.5mg of the

Benznidazole 100mg and also the new indication for children (Appendix 1).

The Brazilian National Regulatory Authority (ANVISA) granted this approval on

December 12, 2011. Brazil is the only country worldwide where Benznidazole 12.5mg

is registered.

The registration number is 25351.111801/2006-44 - 11/2011, and the official

publication in the Brazilian Bulletin can be found at

http://www.in.gov.br/autenticidade.html under the code 10102011121200003

Benznidazole 12.5mg was approved for pediatric use, following years of off label use of

the 100 mg tablet strength for the treatment of children.

Page 57: proposal for the inclusion of benznidazol pediatric dosage

57

14. Availability of pharmacopoeial standards (British Pharmacopoeia, International Pharmacopoeia, United States Pharmacopoeia)

Results from consultations in online search data bases (last update on 26/10/12) are the following:

International Pharmacopoeia / WHO International Chemical Reference Substances (ICRS): Not available

British Pharmacopoeia / British Pharmacopoeia Chemical Reference Substances (BPCRS): Not available

United States Pharmacopeia / USP Reference Standards: Not available

As this standard was never officially available - it includes the Brazilian Pharmacopoeia 5 (2010) which has the monograph of the active principle ingredientcvii (1) -, the manufacturer of the drug product used as reference the Benznidazole Work Standard (WS) assigned by the API manufacturer.

Work is planned in 2013 with US Pharmacopeia to develop USP and WHO reference standards.

15. Proposed (new/adapted) text for the WHO Model Formulary

The adaptation proposed as follows is in the WHO Model Formulary for Children 2010,

in the section American trypanosomiasis (6.5.5.2) for the inclusion of the benznidazole

12.5mg as an additional dosage form. Proposed language in italics was obtained from

the Lafepe package insert for benznidazol 12.5mg and 100mg (Appendix 6 and 7).

There is also a proposal of deletion (in WORD track-changes) in the neonate dose

recommendation on the basis of the new WHO recommendation (2011).

Benznidazole

ATC code: P01CA02

Tablet: 100 mg

Tablet: 12.5 mg

Indications: Treatment of Chagas disease (American trypanosomiasis).

Contraindications: Pregnancy.

This medicine should not be used in case of hypersensitivity to benznidazole or any

other component is in the tablet composition. No health condition is considered an

absolute contraindication to treatment. Benznidazole should only be administered to

pregnant women in cases of an absolute medical indication.

Precautions: Hepatic impairment; renal impairment; haematological conditions;

history of neurological clinical manifestations; allergic condition to imidazoles; monitor

Page 58: proposal for the inclusion of benznidazol pediatric dosage

58

blood count, especially leukocytes, throughout treatment. Benznidazole treatment in

these conditions should be done under medical supervision.

Dose:

Treatment of congenital, acute phase or early chronic phase of Chagas disease

(American trypanosomiasis).

Full-term neonate. Dose must be given in 2–3 divided doses for 60 days.

Treatment of acute phase or early chronic phase of Chagas disease (American

trypanosomiasis).

Oral:

Infant or Child under 40 kg 7.5 mg/kg daily in 2–3 divided doses for 60 days;

40 kg and over 5 mg/kg daily in 2–3 divided doses for 60 days.

NOTE Acute meningoencephalitis may require a dose of up to 25 mg/kg daily.

Treatment of chronic phase of Chagas disease (American trypanosomiasis).

Oral:

Infant or Child 5 mg/kg daily in 2–3 divided doses for 60 days.

NOTE The treating physician should determine the age limits and clinical suitability of

this specific therapy.

In children under twelve years old, especially those in the acute phase of the disease, it

is recommended that doses of 5-10 mg / kg body weight be divided into two daily

doses, for 60 days without interruption.

Summary of the weight categories per dose for tablets Benznidazole 12.5mg

Weight (kg) Recommended Doses (5-10 mg/kg)

Between 2,5kg and <5kg

1 tablet of 12.5mg twice a day for 60 days

(total dose of 25mg/day)

Between 5kg and <10kg

2 tablets of 12.5mg (25 mg) twice a day for

60 days (total dose of 50mg/day)

Between 10kg and <15kg

3 tablets of 12.5 mg (37.5 mg) twice a day

for 60 days (total dose of 75mg/day)

Renal impairment: Avoid use in renal failure (limited data available).

Hepatic impairment: Avoid use in hepatic failure (limited data available).

Adverse effects:

Common Rashes (discontinue treatment if severe and accompanied by fever and

purpura), nausea, vomiting and abdominal pain, peripheral neuropathy.

Uncommon Paraesthesia, peripheral neuritis, leukopenia, arthralgia, myalgia.

Rare Agranulocytosis, bone marrow depression, headache, dizziness, fatigue.

Interactions with other medicines (* indicates severe):

Page 59: proposal for the inclusion of benznidazol pediatric dosage

59

There are no known interactions where it is recommended to avoid concomitant use.

Patients should avoid use of alcohol while using benznidazole.

Concomitant use of benznidazole and aspirin may increase the risk of bleeding.

The effect of anticoagulants such as warfarin may be increased when used together

with benznidazole, due to the inhibition of enzymatic metabolism.

The patient should inform the physician if he/she are taking any other medications.

References: Hill SR, Kouimtzi M, Stuart MC, eds. WHO model formulary. Geneva, World Health Organization, 2008. Kirchhoff LV. Chagas disease (American trypanosomiasis): treatment & medication. eMedicine. New York, WebMD, 2009 (http://emedicine.medscape.com/article/214581-treatment, accessed 10 February 2010). Klasco RK, ed. Drugdex system. Greenwood Village, Thomson Micromedex, 2010. (http://www.thomsonhc.com, accessed 10 February 2010). WHO expert committee on the control of Chagas disease. Control of Chagas disease: second report of the WHO expert committee. WHO Technical Report Series, 2002, 905 (http://whqlibdoc.who.int/trs/WHO_TRS_905.pdf).

List of Appendices Appendix 1- Supporting letter from Brazilian government

Appendix 2 - Supporting letter from Honduras government

Appendix 3 - Supporting letter from Argentinean government

Appendix 4 - Supporting letter from Médecins Sans Frontières

Appendix 5 - ANVISA registration number: 1018301450062

Appendix 6 - Package insert for patient and health professionals

Appendix 7 – Package insert for health professionals

Appendix 8 – Clinical Trial Protocol - Population Pharmacokinetics Study in Children

Page 60: proposal for the inclusion of benznidazol pediatric dosage

60

References

1 World Health Organization (WHO). Working to overcome the global impact of neglected tropical diseases - First WHO report on neglected tropical diseases. 2010. Available at http://whqlibdoc.who.int/publications/2010/9789241564090_eng.pdf

2 International Nonproprietary Names for Pharmaceutical Substances, 1974, Vol.28, No 10, available at

http://whqlibdoc.who.int/inn/INN_1974_list14.pdf 3 Organización Panamericana de la Salud. Estimacion cuantitativa de la enfermedad de Chagas en las

Americas. Montevideo, Uruguay: Organizacion Panamericana de la Salud, 2006 (in Spanish).

4 Arthur M. Strosberg, Ph.D., Kimberly Barrio, M.B.A., Valerie H. Stinger, M.B.A., Jessica Tashker, Ph.D.,

Judith C. Wilbur, Ph.D., Leslie Wilson, Ph.D., and Katherine Woo, Ph.D. CHAGAS DISEASE: A Latin American Nemesis. Institute for One World Health. 2005. Available at: http://www.iowh.net/pdf/CHAGAS%20Landscape%20FINAL%20VERSION.pdf?PHPSESSID=13190f66fe244ed00c40046e1183f20d

5 Organización Panamericana de la Salud. Salud en las Américas. OPS, 2007.

6 World Health Organization (WHO). Working to overcome the global impact of neglected tropical diseases - First WHO report on neglected tropical diseases. 2010. Available at http://whqlibdoc.who.int/publications/2010/9789241564090_eng.pdf

7 Moncayo A, Ortiz Yanine M. An update on Chagas disease (human American trypanosomiasis). Ann

Trop Med Parisitolol. 2006, 100>1-15.

8 Guerri-Guttenberg R, Grana D, Ambrosio G and Milei J. Chagas cardiomyopathy: Europe is not spared!. European Heart Journal 2008- 29, 2587–2591.

9 Gabriel A Schmunis - Epidemiology of Chagas disease in non-endemic countries: the role of international migration - Mem Inst Oswaldo Cruz, Rio de Janeiro, Vol. 102(Suppl. I): 75-85, 2007.

10 http://www.ine.es/prodyser/pubweb/eni07/eni07_anexo.pdf

11 Muñoz, J, et al. Clinical profile of Trypanosoma cruzi infection in a non-endemic setting: Immigration and Chagas disease in Barcelona (Spain). Acta Trop. 2009, doi:10.1016/j.actatropica.2009.02.005.

12 Bern, C. and S.P. Montgomery, An estimate of the burden of Chagas disease in the United States. Clin Infect Dis, 2009;49(5):e52-4.

13 WHO Expert Committee on the Control of Chagas Disease - Brasilia, 20–28 November 2000 – p. 86.

14 Schmunis GA. A Tripanossomiase Americana e seu impacto na saúde publica das Américas. In: Brener Z, Andrade ZA, Barral-Netto M, editors. Trypanosoma cruzi e Doenca de Chagas, 2nd ed. Rio de Janeiro: Guanabara Koogan; 1999. p. 1-15.

15 WHO World Health Report 2002 p. 186- http://www.who.int/whr/2002/en/whr02_en.pdf.

16 Jan H. F. Remme, Piet Feenstra, P. R. Lever, André Médici, Carlos Morel, Mounkaila Noma, K. D.

Ramaiah, Frank Richards, A. Seketeli, Gabriel Schmunis, W. H. van Brakel, and Anna Vassall, "Tropical

Diseases Targeted for Elimination: Chagas Disease, Lymphatic Filariasis, Onchocerciasis, and Leprosy."

2006. Disease Control Priorities in Developing Countries (2nd Edition),ed. , p.436. New York: Oxford

University Press. DOI: 10.1596/978-0-821-36179-5/Chpt-22. http://www.dcp2.org/pubs/DCP.

17 Organizacion Panamericana de la Salud. Estimacion cuantitativa de la enfermedad de Chagas en las Americas. Montevideo, Uruguay: Organizacion Panamericana de la Salud, 2006 (in Spanish).

Page 61: proposal for the inclusion of benznidazol pediatric dosage

61

18

Riera C, Guarro A, El Kassab H, Jorba JM, Castro M, Angrill R, Gallego M, Fisa R, Martin C, Lobato A and Portus M. Congenital transmission of Trypanosoma cruzi in Europe (Spain): A case report - Am. J. Trop. Med. Hyg., 75(6), 2006, pp. 1078–1081.

19 Schijman AG, Altcheh J, Burgos JM, Biancardi M, Bisio M, Levin MJ, Freilij H. Aetiological treatment of

congenital Chagas’ disease diagnosed and monitored by the polymerase chain reaction. J Antimicrob

Chemother. (2003) 52, 441–449.

20 Torrico F, Alonso-Vega C, Suarez E, Rodriguez P, Torrico MC, Dramaix M, Truyens C, Carlier Y. Maternal

Trypanosoma Cruzi Infection, Pregnancy Outcome, Morbidity, and Mortality of Congenitally Infected and

Non-Infected Newborns in Bolivia. Am. J. Trop. Med. Hyg., 70(2), 2004, pp. 201–209

21 Brutus L, Castillo H, Bernal C, Salas NA, Schneider D, Santalla JA, Chippaux JP. Short Report: Detectable

Trypanosoma cruzi Parasitemia during Pregnancy and Delivery as a Risk Factor for Congenital Chagas

Disease. Am J Trop Med Hyg. 2010 Nov;83(5):1044-7.

22 Brutus L, Schneider D, Postigo J, Romero M, Santalla J, Chippaux JP.Congenital Chagas disease:

Diagnostic and clinical aspects in an area without vectorial transmission, Bermejo, Bolívia. Acta Trop.

2008 Jun;106(3):195-9.

23 Carlier Y, Torrico F. Congenital infection with Trypanosoma cruzi: from mechanisms of transmission to strategies for diagnosis and control. Rev Soc Bras Med Trop. 2003 Nov-Dec;36(6):767-71.

24 Rassi A, Amato Neto V, Rassi GG, Amato VS, Rassi Júnior A, Luquetti AO, Rassi SG. Busca retrospectiva

da transmissão maternal da infecção chagásica em pacientes na fase crônica. Rev Soc Bras Med Trop.

2004 Nov-Dec;37(6):485-9

25 Luquetti AO, Ferreira AW, Oliveira RA, Tavares SB, Rassi A, Dias JC, Prata A. Congenital transmission of Trypanosoma cruzi in Brazil: estimation of prevalence based on preliminary data of national serological surveys in children under 5 years old and other sources. Rev Soc Bras Med Trop. 2005;38 Suppl 2:24-6. Rev Soc Bras Med Trop. 2005;38 Suppl 2:24-6.

26 Wegner DH, Rohwedder RW. The effect of nifurtimox in acute Chagas’ infection.

Arzneimittelforschung. 1972;22(9):1624-1635.

27 Kirchhoff LV. Chagas disease: American trypanosomiasis. Infect Dis Clin North Am. 1993; 7(3):487-502.

28 Cancado JR, Brener Z. Terapeutica. In: Brener Z, Andrade Z, eds. Trypanosoma cruzi e doenca de

Chagas.Rio de Janeiro, Brazil: Guanabara Koogan; 1979: 362-424.

29 FERREIRA, H.O. - Tratamento específico na fase aguda da doença de Chagas. J. Pediat., 64: 126-128,

1988.

30 FERREIRA, H.O. - Tratamento da forma indeterminada da doença de Chagas com nifurtimox e benzonidazol. Rev. Soc. bras. Med. trop., 23: 209-211, 1990.

31 Barclay, C.A., Cerisola, J.A., Lugones, H., Ledesma, O., Silva, J.L., Mouzo, G. Aspectos farmacológicos y resultados terapêuticos del benznidazol en el tratamiento de la infección chagásica. Prensa. Med. Arg. 65: 239-244, 1978.

32 Russomando G, de Tomassone MM, de Guillen I, Acosta N, Vera N, Almiron M, et al. Treatment of congenital Chagas' disease diagnosed and followed up by the polymerase chain reaction. Am J Trop Med Hyg. 1998 Sep;59(3):487-91.

33 Russomando G, Almirón M, Candia N, Franco L, Sánchez Z, de Guillen I. Implementation and

evaluation of a locally sustainable system of prenatal diagnosis to detect cases of congenital Chagas

Page 62: proposal for the inclusion of benznidazol pediatric dosage

62

disease in endemic areas of Paraguay. Rev Soc Bras Med Trop. 2005;38 Suppl 2:49-54.

34 Cançado JR 2002. Long term evaluation of etiological treatment of Chagas disease with benznidazol.

Rev Inst Med Trop Sao Paulo 44: 20-37.

35 Torrico F, Alonso-Vega C, Suarez E, Rodriguez P, Torrico MC, Dramaix M, Truyens C, Carlier Y. Maternal

Trypanosoma Cruzi Infection, Pregnancy Outcome, Morbidity, and Mortality of Congenitally Infected and Non-Infected Newborns in Bolivia. Am. J. Trop. Med. Hyg., 70(2), 2004, pp. 201–209.

36 Salas NA, Cot M, Schneider D, Mendoza B, Santalla JA, Postigo J, Chippaux JP, Brutus L. Risk factors and consequences of congenital Chagas disease in Yacuiba, south Bolívia. Trop Med Int Health. 2007 Dec;12(12):1498-505.

37 Chippaux JP, Clavijo AN, Santalla JA, Postigo JR, Schneider D, Brutus L. Antibody drop in newborns congenitally infected by Trypanosoma cruzi treated with benznidazole. Trop Med Int Health. 2010 Jan;15(1):87-93.

38 de Andrade ALS, Zicker F, de Oliveira RM, Almeida Silva S, Luquetti A, Travassos LR, Almeida IC, de

Andrade SS, de Andrade JG, Martelli CM 1996. Randomosed trial of efficacy of benznidazole in treatment of early Trypanosoma cruzi infection. Lancet 348: 1407-1413

39 Sosa-Estani S, Segura EL, Ruiz AM, Velazquez E, Porcel BM, Yampotis C 1998. Chemotherapy with

benznidazole in children in undetermined phase of Chagas disease. Am J Trop Med Hyg 59: 526-529.

40 Sosa-Estani S, Herrera de Bravo B, Herrera de Bizzoto L, Canil S, Cura EN, Segura EL 2002. Evolución serológica a largo plazo en niños infectados por Trypanosoma cruzi que cursan fase clínica indeterminada, tratados con benznidazol. Available at: http://www.fac.org.ar/fec/chagas2/llave/md8/md804/sosaes.htm, 2002.

41 Flores-Chavez M, Bosseno MF, Bastrenta B, Alcazar Dalenz JL, Hontebeyrie M, Revollo S, Brenière SF 2006. Polymerase Chain Reaction Detection and Serologic Follow-Up after Treatment with Benznidazole in Bolivian Children Infected with a Natural Mixture of Trypanosoma Cruzi I And Ii. Am. J. Trop. Med. Hyg., 75(3),. 497–501.

42 Duffy T, Bisio M, Altcheh J, Burgos JM, Diez M, Levin MJ, Favaloro RR, Freilij H, Schijman AG. Accurate real-time PCR strategy for monitoring bloodstream parasitic loads in chagas disease patients. PLoS Negl Trop Dis. 2009;3(4):e419.

43 Blanco SB, Segura EL, Cura EN, Chuit R, Tulián L, Flores I, Garbarino G, Villalonga JF, Gürtler RE. Congenital transmission of Trypanosoma cruzi: an operational outline for detecting and treating infected infants in north-western Argentina. Trop Med Int Health. 2000 Apr;5(4):293-301.

44 Silveira CAN, Castillo E, Castro C 2000. Avaliação do tratamento específico para o Trypanosoma cruzi

em crianças, na evolução da fase indeterminada. Rev Soc Bras Med Trop. 33(2):191-196.

45 Streiger ML, del Barco ML, Fabbro DL, Arias ED, Amicone NA 2004. Longitudinal study and specific

chemotherapy in children with chronic Chagas’ disease, residing in a low endemicity area of Argentina. Rev Soc Bras Med Trop 37: 365-375.

46 Rodriques Coura J, deCastro SL. A critical review on Chagas disease chemotherapy. Mem Inst

OswaldoCruz. 2002;97(1):3-24.

47 Altcheh J, Biancardi M, Lapena A, Ballering G, Freilij H. Congenital Chagas disease: experience in the

Hospital de Niños, Ricardo Gutıerrez, Buenos Aires, Argentina [in Spanish]. Rev Soc Bras Med Trop. 2005; 38(suppl 2):41-45.

48 Secretaria de Vigilância em Saúde. Guia de Vigilância Epidemiológica. 7ª Edição. Available at:

http://portal.saude.gov.br/portal/arquivos/pdf/gve_7ed_web_atual_doenca_de_chagas.pdf.

49 World Health Assembly, 2010. WHA 63.20 - Chagas disease: control and elimination.

Page 63: proposal for the inclusion of benznidazol pediatric dosage

63

50 Carlier Y, Torrico F, Sosa-Estani S, Russomando G, Luquetti A, et al. (2011) Congenital Chagas Disease: Recommendations for Diagnosis, Treatment and Control of Newborns, Siblings and Pregnant Women. PLoS Negl Trop Dis 5(10): e1250. doi:10.1371/journal.pntd.0001250

51 Jackson Y, Myers C, Diana A, Marti HP, Wolff H, Chappuis F, Loutan L, and Gervaix A. Congenital Transmission of Chagas Disease in Latin American Immigrants in Switzerland. Emerging Infectious Diseases, Vol. 15, No. 4, April 2009: 601-603.

52 Basile L, Oliveira I, Ciruela P, Plasencia A, working group for developing the Catalonian Screening Programme for congenital transmission of Chagas disease. The current screening programme for congenital transmission of Chagas disease in Catalonia, Spain. Spain. Euro Surveill. 2011;16(38):pii=19972. Available online: http://www.eurosurveillance.org/ViewArticle.aspx?ArticleId=19972

53 Centers for Disease Control and Prevention. Congenital Transmission of Chagas Disease — Virginia,

2010. MMWR 2012;61 (26):477-479.

54 Teng J, Song CK, Williams RL, Polli JE. Lack of medication dose uniformity in commonly split tablets. J Am Pharm Assoc (Wash). 2002 Mar-Apr;42(2):195-9.

55 WHO. Control of Chagas Disease: Second Report of the WHO Expert Committee. WHO Technical

Report Series, No. 905. . Geneva: World Health Organization; 2002.

56 Raaflaub J. Multiple-dose kinetics of the trypanosomide benznidazole in man. . Arzneimittelforschung. 1980;30(12):2192-4.

57 Raaflaub J ZW. Single-dose pharmacokinetics of the trypanosomide benznidazole in man. Arzneimittelforschung. 1979;29:1611-4.

58 WHO 2003. Scaling up antiretroviral therapy in resource limited settings: Treatment Guidelines for a Public Health Approach – 2003 revision, WHO-Geneva.

59 República Federativa do Brasil. Agência Nacional de Vigilância Sanitária. RESOLUÇÃO-RDC No- 48, DE 6

DE OUTUBRO DE 2009. Available at:

http://portal.anvisa.gov.br/wps/wcm/connect/ad719600419766e68cb0ad925ac4fc61/RDC_48_2009_P

os_registro.pdf?MOD=AJPERES.

60 Brasil. Ministério da Saúde. Relação Nacional de Medicamentos Essenciais RENAME 2010. 7ª Edição.

Brasília, 2010. Available at http://portal.saude.gov.br/portal/arquivos/pdf/rename2010final.pdf.

61 Brasil. Ministério da Saúde. PORTARIA Nº 1.284, DE 26 DE MAIO DE 2010. Available at: http://www.brasilsus.com.br/legislacoes/gm/104209-1284.

62 Relação Nacional de Medicamentos Essenciais – RENAME, 2012. Available at

http://portal.saude.gov.br/portal/arquivos/pdf/CONITECANEXOSRENAME.pdf (page 48)

63 World Health Organization. Control of Chagas Disease: Second Report of the WHO Expert Committee.

WHO Technical Report Series, No. 905. 2002. Geneva: World Health Organization.

64 WHO Drug Information Vol. 26, No. 1, 2012, page 21. Benznidazole: child-adapted dosage form

approved. Available at http://www.who.int/medicines/publications/druginformation/issues/26-1.pdf

65 IRD – 2009- L. Brutus (attachment) – personal communication (report from Chagas Clinical Trial Platform)

Page 64: proposal for the inclusion of benznidazol pediatric dosage

64

66

Galvão LM, Chiari E, Macedo AM, Luquetti AO, Silva SA, Andrade AL. PCR Assay for Monitoring

Trypanosoma cruzi Parasitemia in Childhood after Specific Chemotherapy. J Clin Microbiol. 2003

Nov;41(11):5066-70.

67 Andrade AL, Martelli CM, Oliveira RM, Silva SA, Aires AI, Soussumi LM, Covas DT, Silva LS, Andrade JG,

Travassos LR, Almeida IC. Short Report: Benznidazole Efficacy among Trypanosoma Cruzi−Infected

Adolescents after a Six-Year Follow-Up. Am J Trop Med Hyg. 2004 Nov;71(5):594-7.

68 Yun O, Lima MA, Ellman T, Chambi W, Castillo S, Flevaud L, Roddy P, Parreño F, Albajar Viñas P, Palma

PP. Feasibility, drug safety, and effectiveness of etiological treatment programs for Chagas disease in

Honduras, Guatemala, and Bolivia: 10-year experience of Médecins Sans Frontières. PLoS Negl Trop Dis.

2009 Jul 7;3(7):e488

69 Escribà JM, Ponce E, Romero Ade D, Albajar Viñas P, Marchiol A, Bassets G, Palma PP, Lima MA, Zúniga

C, Ponce C 2009 . Treatment and seroconversion in a cohort of children suffering from recent chronic

Chagas infection in Yoro, Honduras. Mem Inst Oswaldo Cruz, Rio de Janeiro, Vol. 104(7): 986-991.

70 Streiger, ML; Barco,ML; Fabbro, DL, Arias,HD e Amicone,NA. Estudo longitudinal e quimioterapia específica em crianças, com doença de Chagas crônica, residentes em área de baixa endemicidade da República Argentina. Revista da Sociedade Brasileira de Medicina Tropical 37(5):365-375, set-out, 2004. 71 Sánchez Negrette O, Sánchez Valdéz FJ, Lacunza CD, García Bustos MF, Mora MC, Uncos AD,

Basombrío MA. Serological Evaluation of Specific-Antibody Levels in Patients Treated for Chronic

Chagas’ Disease. Clin Vaccine Immunol. 2008 Feb;15(2):297-302.

72 Viotti R, Vigliano C, Armenti H, Segura E. Treatment of chronic Chagas’ disease with Bze: Clinical and

serologic evolution of patients with long- term follow - up. Am Heart J 1994; 127:151-162.

73 Pinazo MJ, Muñoz J, Posada E, López-Chejade P, Gállego M, Ayala E, del Cacho E, Soy D, Gascon J. Tolerance of benznidazole in treatment of Chagas' disease in adults. Antimicrob Agents Chemother. 2010 Nov;54(11):4896-9

74 Coura JR, de Abreu L, Willcox HP, Petana W 1997. Comparative controlled study on the use of benznidazole, nifurtimox and placebo, in the chronic form of Chagas' disease in a field area with interrupted transmission. I. Preliminary evaluation. Rev Soc Bras Med Trop 30: 139-144. 75 Viotti, R; Vigliano, C; Lococo, B; Bertocchi, G; Petti, M; Alvarez, MG; Postan, M; and Armenti, A. Long-

Term Cardiac Outcomes of Treating Chronic Chagas Disease with Benznidazole versus No Treatment -A

Nonrandomized Trial. Ann Intern Med. 2006;144:724-734. 76 Gallerano RR, Sosa RR 2000. Estudio de intervención en la evolución natural de la enfermedad de Chagas. Evaluación del tratamiento antiparasitario específico. Estudio retrospectivo-prospectivo de terapéutica antiparasitaria. Rev Fac Cienc Med Cordoba 57: 135-162. 77 Sosa-Estani S, Armenti A, Araújo G, Viotti R, Lococo B, Ruiz Vera B, Vigliano C, De Rissio AM, Segura EL. Tratamiento de la enfermedad de Chagas con benznidazol y ácido tioctico. Medicina (Buenos Aires) 2004; 64:1-6.

78 de Pontes VM, Souza Júnior AS, Cruz FM, Coelho HL, Dias AT, Coêlho IC, Oliveira Mde F. Reações

adversas em pacientes com doença de Chagas tratados com benzonidazol, no Estado do Ceará. Rev Soc

Bras Med Trop. 2010 Apr;43(2):182-7.

79 Levi GC, Lobo IM, Kallás EG, Amato Neto V. Etiological drug treatment of human infection by Trypanosoma cruzi.Rev Inst Med Trop Sao Paulo. 1996 Jan-Feb;38(1):35-8.

Page 65: proposal for the inclusion of benznidazol pediatric dosage

65

80 Cadernos de Atenção Básica. Vigilância em Saúde. Zoonoses. Ministério da Saúde. Secretaria de Atenção à Saúde. Departamento de Atenção Básica. Brasília, 2009. Disponível em: http://portal.saude.gov.br/portal/arquivos/pdf/abcad22.pdf

81 Secretaria de Vigilância em Saúde do Ministério da Saúde. Consenso Brasileiro em Doença de Chagas.

Rev Soc Bras Med Trop. Vol. 38 (Suplemento III), 2005. Available at:

portal.saude.gov.br/portal/arquivos/pdf/consenso_chagas.pdf

82 Amato Neto V, Lopes MH. Carta ao Editor. Revista da Sociedade Brasileira de Medicina Tropical 34(2):

221, mar-abr, 2001.

83 Moll C, Peris P, Moreno A, Muñoz J, Guañabens N. Severe invalidating pain syndrome associated with benznidazole therapy for Chagas' disease .Clin Rheumatol. 2008 Feb;27(2):269-70.

84 Castro JA, de Mecca MM, Bartel LC. Toxic Side Effects of Drugs Used to Treat Chagas' Disease (American Trypanosomiasis). Hum Exp Toxicol. 2006 Aug;25(8):471-9. 85 Bartel, L.C., Montalto de Mecca, M., et al., 2007. Early nifurtimox-induced biochemical and ultrastructural alterations in rat heart. Hum. Exp. Toxicol. 26 (10), 781–788.

86 de Toranzo EGD, Masana M, Castro JA. Administration of Benznidazole, a chemotherapeutic agent

against Chagas’ disease to pregnant rats. Covalent binding of reactive metabolites to fetal and maternal

proteins. Arch Int Pharmacodyn Ther 1984; 272: 17_23.

87 de Mecca, M.M., Bartel, L.C., et al., 2008. Benznidazole biotransformation in rat heart microsomal fraction without observable ultrastructural alterations: comparison to Nifurtimox-induced cardiac effects. Mem. Inst. Oswaldo Cruz 103 (6), 549–553.

88 de Castro CR, de Toranzo EGD, Castro JA. Benznidazole- induced ultrastructural alterations in rat

adrenal cortex. mechanistic studios. Toxicology 1992; 74: 223_ 32.

89 Diaz EGD, de Castro CR, de Mecca MM, Castro JA. Benznidazole-induced ultrastructural and

biochemical alterations in the rat colon. Acta Pharmacol Sin 2000; 21: 961_66.

90 de Castro CR, de Mecca MM, Fanelli SL, de Ferreyra EC, Diaz EG, Castro JA. Benznidazole-induced

ultrastructural and biochemical alterations in rat esophagus. Toxicology 2003; 191: 189_98.

91 Navarro ML, Dain L, Miglionni AM, Nagel R. Clastogenic activity of two antichagasic drugs.

Comunicaciones Biológicas 1984; 3: 25_28.

92 R. C. C. Ferreira, M. E. B. de Melo, M. A. Moraes Jr., and L. C. S. Ferreira, “Evaluation of genotoxic

activity in the blood and urine of guinea pigs treated with nifurtimox and benznidazole,” Brazilian

Journal of Medical and Biological Research, vol. 21, no. 5, pp. 1069–1077, 1988.

93 N. B. Gorla. Sister-chromatid exchange in spleenic lymphocytes of mice after exposure to nifurtimox

or benznidazole. Mutation Research, vol. 188, no. 2, pp. 129–133, 1987.

94 Buschini A,Ferrarini L, Franzoni S, Galati S, Lazzaretti M, Mussi F, Northfleet de Albuquerque C, Maria

Araújo Domingues Zucchi T, Poli P. Genotoxicity revaluation of three commercial nitroheterocyclic

drugs: nifurtimox, benznidazole, and metronidazole. J Parasitol Res. 2009;2009:463575.

95 Gorla NB, Ledesma OS, Barbieri GP, Larripa IB. Assessment of cytogenetic damage in chagasic children treated with benznidazole. Mutat Res. 1988 Oct;206(2):217-20.

Page 66: proposal for the inclusion of benznidazol pediatric dosage

66

96

Gorla NB, Castro JA. Micronucleus formation in bone marrow of mice treated with Nifurtimox or

Benznidazole. Toxicol Lett 1985; 25: 259_63.

97 S. C. Souza, C. S. Takahashi, and J. S. da Silva, “Evaluation of themutagenic potential of the

antichagasic drug Rochagan in healthy and Chagasic rodents,” Mutation Research, vol. 259, no. 2, pp.

139–145, 1991.

98 Sosa Estani. Therapy, diagnosis and prognosis of chronic Chagas disease: insight gained in Argentina. Mem Inst Oswaldo Cruz, Rio de Janeiro, Vol. 104(Suppl. I): 167-180, 2009.

99 Teixeira AR, Calixto MA, Teixeira ML. Chagas’ disease, carcinogenic activity of the anti trypanosomal

nitrioarenes in mice. Mutat Res 1994; 305: 189_96.

100 Teixeira AR, Silva R, Cunha Neto E, Santana JM, Rizzo LV 1990. Malignant, non-Hodgkin’s lymphomas

in Tripanosoma cruzi-infected rabbits treated with nitroarenes. J Comp Pathol 103: 37-48.

101 Aguilar EG, Arranz CK, de Toranzo EGD, Castro, JA. Liver microsomal Benznidazole and Nifurtimox

nitroreductase activity in male rats of different age. Arch Int Pharmacodyn Ther 1987; 289: 11_17.

102 de Toranzo EGD, Castro JA, de Cazzulo BMF, Cazzulo JJ. Interaction of Benznidazole reactive

metabolites with nuclear and kinetoplastic DNA; proteins and lipids from Trypanosoma cruzi.

Experientia 1988; 44: 880_81.

103 Altcheh J, Moscatelli G, Moroni S, Garcia-Bournissen F, Freilij, H. Adverse Events After the Use of

Benznidazole in Infants and Children With Chagas Disease. Manuscrito aceito para publicação em

Pediatrics 2011;127 (1).

civ World Health Organization. Accelerating work to overcome the global impact of neglected tropical diseases - a roadmap for implementation. Published in 2012. Reference to donnation programm in page 15. Available at http://www.who.int/neglected_diseases/NTD_RoadMap_2012_Fullversion.pdf

cv Sicuri E, Muñoz J, Pinazo MJ, Posada E, Sanchez J, Alonso PL, et al. Economic evaluation of Chagas disease screening of pregnant Latin American women and of their infants in a non-endemic area.Acta Trop. 2011;118(2):110-7.

cvi Billot, C., Torrico, F., Carlier, Y., 2005. Cost effectiveness study of a control program of congenital Chagas disease in Bolivia. Rev. Soc. Bras. Med. Trop. 38 (Suppl. 2), 108–113 cvii Available as "BENZNIDAZOL" on pages 683-684 at

http://www.anvisa.gov.br/hotsite/cd_farmacopeia/pdf/volume2.pdf