Mathematical modelling of disease progression

Post on 22-Jan-2018

857 Views

Category:

Health & Medicine

2 Downloads

Preview:

Click to see full reader

Transcript

A mechanism-based disease progression model to analyse long-term treatment

effects on disease processes underlying type 2 diabetes

Workshop“The interplay of fat and carbohydrate metabolism with application in Metabolic Syndrome and Type 2

Diabetes”

December 12th 2013

Yvonne Rozendaaly.j.w.rozendaal@tue.nl

Introduction

• Disease progression– multi-scale problem

– how to assess/measure?

• Treatment interventions– effect of treatment on disease progression?

short-term vs long-term

• How to simulate adaptations & interventions?

2

Type 2 Diabetes Mellitus (T2DM)

• Impaired beta-cell function

• Reduced insulin sensitivity

• Monitoring glycemic control: biomarkers

– FPG: fasting plasma glucose

– FSI: fasting serum insulin

– HbA1c: glycosylated hemoglobin

3

chronic loss of glycemic control

secondaryglycemic markers

primary glycemic marker

how to derivedisease status?

T2DM treatment

• hypoglycemic effect: short-term

– immediate symptomatic effects on glycemiccontrol

• inhibitory effect on disease progression: long-term

– protect against T2DM progression

4

Objective

5

metabolic biomarkersFPGFSI

HbA1c

treatment interventionspharmacological therapy

disease progressionprogressive loss of beta-cell

function and insulin sensitivityadaptations in

biological network

disease progression model introduction to ADAPT application of ADAPT

computational model:description and quantification of inputs

test functionality of method on minimal model: human vs. mouse glucose vs. lipid metabolism

minimal model

• Disentangle treatment effects– long-term

loss of beta-cell functionand insulin sensitivity

– short-termanti-hyperglycemic effects

• Computational model:study & quantifytime-course effects

6

de Winter et al. (2006) J Pharmacokinet Pharmacodyn,33(3):313-343

Modelling disease progression (1)

disease progression model introduction to ADAPT application of ADAPT

PK/PD modelling

• PharmacoKinetic-PharmacoDynamic modelling

• Simple kinetics are modelled using minimal/macroscopic models

• e.g. absorption profiles

7disease progression model introduction to ADAPT application of ADAPT

T2DM disease progression model (1)glucose – insulin – HbA1c

• Model components– FPG: fasting plasma glucose

– FSI: fasting serum insulin

– HbA1c: glycosylated hemoglobin

• Physiological FPG-FSI homeostasis:– feedback between FSI and FPG

FPG stimulates FSI production: FSI production rate ∝ FPG concentration

– feed-forward between FPG and HbA1cHbA1c production rate ∝ to FSI concentration

8disease progression model introduction to ADAPT application of ADAPT

9

ink

ink

ink

outk

outk

outk

B: beta-cell function(disease status)

S: insulin sensitivity(disease status)

FPG

HbA1c

FSI

EFS: insulin sensitizingeffect of treatment

EFB: treatment effecton insulin secretion

feed-forward

homeostaticfeed-backs

T2DM disease progression model (2)model structure

disease progression model introduction to ADAPT application of ADAPT

10

1cHbA1cHbA

FPG

FPG

FSIFSI

1c

1c HbAFPGt

HbA

FPGFSIt

FPG

FSI)5.3FPG(t

FSI

outin

out

S

in

outinB

kkd

d

kSEF

k

d

d

kkBEFd

d

disease status:fraction of remaining beta-cell function

disease status:fraction of remaining insulin sensitivity

treatment specific factor of insulin-

sensitizers

treatment specific factor of insulin-

secretogogues

T2DM disease progression model (3)model equations

disease progression model introduction to ADAPT application of ADAPT

• Beta-cell functionfraction of remainingbeta-cell function

• Insulin sensitivityfraction of remaininghepatic insulin-sensitivity

• Assumption: asympotically decrease over time

11

)exp(1

1

0trb

B

B

)exp(1

1

0trs

S

S

shift of disease progression curve

slope of disease

progression curve

T2DM disease progression model (1)disease status

disease progression model introduction to ADAPT application of ADAPT

Model comparison with data (1)

• Long-term (1y) follow-up of treatment-naïve T2DM patients

• 3 treatment arms: monotherapy with different hypoglycemic agents– pioglitazone: insulin sensitizer

• enhances peripheral glucose uptake• reduces hepatic glucose production

– metformin: insulin sensitizer• decreases hepatic glucose production

– gliclazide: insulin secretogogue• stimulates insulin secretion by the pancreatic beta-cells

12disease progression model introduction to ADAPT application of ADAPT

Model comparison with data (2)

13

FPG

[m

mo

l/L]

disease progression model introduction to ADAPT application of ADAPT

Reproduction of results (1)

14

Metabolic biomarkers over time

although initial decrease, glycemiccontrol still gradually decreases over time

disease progression model introduction to ADAPT application of ADAPT

Reproduction of results (2)

15

Disease status

however, morphology of disease progression curves unknown...

gliclazide:insulin secretogogue

pioglitazone & metformin:insulin sensitizers

disease progression model introduction to ADAPT application of ADAPT

Introduction to ADAPT (1)

• Phenotype transition over time

• Analysis of Dynamic Adaptations in Parameter Trajectories

16

treatment interventionsmedication, surgery, ... disease progression

which adaptations occur?

Tiemann et al. (2011). BMC Syst Biol,26(5):174Tiemann et al. (2013). PLoS Comput Biol,9(8):e1003166

phenotype A phenotype B

disease progression model introduction to ADAPT application of ADAPT

Introduction to ADAPT (2)

• Phenotype transition:– gradual, long-term processes– measurements at metabolome level

• Adaptation at proteome and transcriptome level

• Model at metabolome level

• Time-dependency implemented using time-varying parameters

17disease progression model introduction to ADAPT application of ADAPT

Modelling phenotype transition (1)

18

treatment

disease progression

long-term discrete data: different phenotypes

disease progression model introduction to ADAPT application of ADAPT

Modelling phenotype transition (2)

19

long-term discrete data: different phenotypes estimate continuous data: cubic smooth spline

introduce artificialintermediate phenotypes

disease progression model introduction to ADAPT application of ADAPT

Modelling phenotype transition (3)

20

long-term discrete data: different phenotypes estimate continuous data: cubic smooth spline incorporate uncertainty in data: multiple describing functions

disease progression model introduction to ADAPT application of ADAPT

Parameter estimation (1)

21

steady state model

disease progression model introduction to ADAPT application of ADAPT

Parameter estimation (2)

22

steady state model iteratively calibrate model to data: estimate parameters over time

minimize difference between data and model simulation

disease progression model introduction to ADAPT application of ADAPT

Parameter estimation (2)

23

steady state model iteratively calibrate model to data: estimate parameters over time

disease progression model introduction to ADAPT application of ADAPT

Parameter estimation (2)

24

steady state model iteratively calibrate model to data: estimate parameters over time

disease progression model introduction to ADAPT application of ADAPT

Parameter estimation (2)

25

steady state model iteratively calibrate model to data: estimate parameters over time

disease progression model introduction to ADAPT application of ADAPT

Estimated parameter trajectories

26

up-regulation

down-regulation

unaffectedstochastic

behaviour...

effect of parameter adaptations on underlying processes?

physiologically unrealistic

disease progression model introduction to ADAPT application of ADAPT

Possible applications for ADAPT

27

• Unravel which processes in network might be responsible for phenotype transition

• Guide new experiment design

• Define possible pharmacological targets

disease progression model introduction to ADAPT application of ADAPT

Application of ADAPT indisease progression model

28

1cHbA1cHbA

FPG

FPG

FSIFSI

HbA1cFPGt

HbA

FPGFSIt

FPG

FSI)5.3FPG(t

FSI

1c

outin

out

in

outin

kkd

d

kS

k

d

d

kkBd

d

fraction of beta-cell function:time-dependent parameter

fraction of insulin sensitivity:time-dependent parameter

time-constantparameters

disease progression model introduction to ADAPT application of ADAPT

29

Metabolic biomarkers over timetreatment with pioglitazone

Disease progression modelvs. application of ADAPT (1)

disease progression model introduction to ADAPT application of ADAPT

HbA1c:performance ADAPT

FPG & FSI:ADAPT reproduces model predictions

30

Parameter trajectories: disease statustreatment with pioglitazone

Disease progression modelvs. application of ADAPT (2)

disease progression model introduction to ADAPT application of ADAPT

ADAPT suggests dynamic disease progression curves rather than pre-defined mathematical functions by de Winter et al.

Disease progression modelvs. application of ADAPT (2)

31

Parameter trajectories: disease statustreatment with pioglitazone

disease progression model introduction to ADAPT application of ADAPT

ADAPT suggests dynamic disease progression curves rather than pre-defined mathematical functions by de Winter et al.

Conclusions & Future work

• Disease progression model & ADAPT approach both useful for monitoring disease status

• ADAPT– applicable to both mice/human, glucose/lipoprotein

metabolism and multiscale models– more dynamically correct representation of beta-cell

function and insulin sensitivity using ADAPT

• However;– How to disentangle disease progression effects from hypoglycemic effects?– How to estimate time-varying parameters in conjunction with time-constant

parameters?

32

Acknowledgements

top related