Background of NHE-1 inhibition in DMD Fibrosis and Inflammation … · 2016. 10. 5. · PK profile...

1
Translational Development of Rimeporide, a Sodium-Hydrogen Exchanger (NHE-1) Inhibitor, for Patients with Duchenne Muscular Dystrophy F. Porte-Thomé 1 , J.B. Su 2 , I. Barthelemy 3 , N.Blanchard Gutton 3 , C. Laveille 4 , G. Bkaily 5 , Q. Yu 6 , K. Nagaraju 6 , B. Ghaleh 2 , S. Blot 3 1 EspeRare Foundation, Geneva, Switzerland ([email protected]); 2 INSERM, Unité 955, Equipe 3, Créteil France; 3 Ecole Nationale Vétérinaire d’Alfort Laboratoire de Neurobiologie Maisons-Alfort France & IMRB, INSERM, Unité 955, Equipe 10, Créteil France; 4 Calvagone, France, 5 University of Medicine, Sherbrooke, Canada, 6 Children’s National Medical Center, Washington DC, USA. Background and rationale Significant reduction of fibrosis in the heart (above, -29%) & in diaphragm (-19%) , in mdx mice after a 9 M treatment) Phase II/III prerequisites 0 25 50 75 100 125 150 175 200 225 250 275 300 325 350 375 400 0.00 0.25 0.50 0.75 1.00 340 jours 100 30 jours AGE (Jours) Survie (%) Prevention of early death in cardiomyopathic hamsters Fibrosis and Inflammation in mdx mice BL10 Vehicle Mdx Vehicle Mdx-400ppm Mdx-800ppm + - Photon Intensity Significant reduction in inflammation in a large panel of skeletal muscles (forelimb & hindlimb) & diaphragm (-38%) Cardioprotection in CMH Hamsters Placebo GMRD moderate GMRD severe GLP Toxicology studies Rimeporide Clinical trial status update A complete safety & PK in healthy adults and congestive heart failure patients Ongoing Clinical development in patients with DMD (6 to 14 y) 2015 2016 2017 2021 Acknowledgements to patients & families , investigators and staff & patients organisations In cohort 1: 5 ambulant DMD patients aged 6 to 10 y, enrolled in France, received oral rimeporide for 4 W No treatment-related adverse events, no significant modification of safety lab & tolerability issues In cohort 2: 5 patients (6-10 y) in Italy, Spain & UK completed, safety analysis ongoing PK: Visual Predictive Check shows that the adult model, incorporating allometric scaling, adequately reflects the PK in children Clinical pharmacology studies in over 166 healthy and CHF patients: Single oral and IV administration Multiple oral doses administration Food interaction Drug Drug Interaction with Digoxin Safety, tolerability, PK in CHF patients with renal insufficiency Adverse events were mild in intensity with no clear dose dependency headache (15%), dizziness ( 5%), chest discomfort (10%), paresthesias, vaso-vagal attacks, local skin reactions. No clinically relevant changes in vital signs, ECGs, Laboratory. Similar events reported on Placebo and treated patients PK profile in Adults Predicted PK profile in Children Primary objective: Determine safety & tolerability of Rimeporide after 4 weeks oral treatment Secondary objective : Evaluate the PK profile of rimeporide in plasma Exploratory objectives: NMRI indices (T2, Muscle Mass, FF) & Serum Biomarkers 0 500 1000 1500 2000 2500 3000 0 6 12 18 24 30 36 42 48 54 60 66 72 EMD 87 580 plasma concentration [ng/mL] . mean 50 mg day1 mean 100mg day1 mean 200 mg day1 mean 50 mg day10 mean 100 mg day10 mean 200 mg day10 0 5 10 15 20 0 500 1000 1500 Rimeporide (µg/L) AGE=6yr AGE=12yr Extensive Toxicology package did show that Rimeporide was safe Repeated oral toxicity studies up to 26-week in rats and up to 39-week in dogs showed the main target organ in both species to be fundic parietal cells (reversible mild parietal cells alteration) No mutagenic activity nor teratogenic potential were shown No impairment of male and female fertility No skin sensitising properties No effect on cardiovascular hemodynamic parameters, no evidence of risk for any pro-arrhythmic effects No relevant effects on the central and peripheral nervous system, nor on the gastro-intestinal, renal and respiratory system Rimeporide [Na + ] i serves as a co-regulator of Ca 2+ influx through the NCX and NHE-1, (Burr et al, 2015) [Na + ] i overload was observed in DMD patients (Weber at al, 2012), is responsible for edema and a secondary increase in basal Ca 2+ levels leading to myofiber necrosis and muscles degeneration (Burr et al, 2015) NHE-1 inhibitors reduce [Na+] i , & indirectly [Ca2+] i (Iwata et al, 2007; Dorchies et al, 2015), reduce myocardial necrosis (Chahine et al, 2005), myofiber fibrosis (Nagaraju et al, 2015), inflammation (Yang et al 2013) and prevent early death (Bkaily et al, 2015) in CM hamsters . Rimeporide - Background of NHE-1 inhibition in DMD Translational GRMD Dogs studies Cohort 1 PK results Decreased heart necrosis, thrombosis, myolysis 20 animals (8 severe & 12 moderate) Blinded treatment with Rimeporide at 20 mg/kg/d or placebo started from 2 to 12 months Objectives: Contribute to the design of the clinical study by guiding dose selection, identifying novel non invasive biomarkers as well as cardiac outcome measures. GMRD moderate Simulated values in Children Observed values in children

Transcript of Background of NHE-1 inhibition in DMD Fibrosis and Inflammation … · 2016. 10. 5. · PK profile...

Page 1: Background of NHE-1 inhibition in DMD Fibrosis and Inflammation … · 2016. 10. 5. · PK profile in Adults Predicted PK profile in Children ... (Chahine et al, 2005), myofiber fibrosis

Translational Development of Rimeporide, a Sodium-Hydrogen Exchanger (NHE-1) Inhibitor, for Patients with Duchenne Muscular Dystrophy

F. Porte-Thomé1, J.B. Su2, I. Barthelemy3 , N.Blanchard Gutton3, C. Laveille4, G. Bkaily5, Q. Yu6, K. Nagaraju6, B. Ghaleh2, S. Blot3

1EspeRare Foundation, Geneva, Switzerland ([email protected]); 2INSERM, Unité 955, Equipe 3, Créteil France; 3Ecole Nationale Vétérinaire d’Alfort Laboratoire de Neurobiologie Maisons-Alfort France & IMRB, INSERM, Unité 955, Equipe 10, Créteil France; 4Calvagone, France, 5University of Medicine, Sherbrooke, Canada, 6Children’s National Medical Center, Washington DC, USA.

Background and rationale

Significant reduction of fibrosis in the heart (above, -29%) & in diaphragm (-19%) , in mdx mice after a 9 M treatment)

Phase II/III prerequisites

0 25 50 75 100 125 150 175 200 225 250 275 300 325 350 375 400

0.00

0.25

0.50

0.75

1.00

Control EMD

EMD 87580

340 jours

100

30 jours

AGE (Jours)

Su

rv

ie

(%)

Prevention of early death in cardiomyopathic hamsters

Fibrosis and Inflammation in mdx mice

BL10 Vehicle Mdx Vehicle Mdx-400ppm Mdx-800ppm

+

-Photon

Intensity

Significant reduction in inflammation in a large panel of skeletal muscles (forelimb & hindlimb) & diaphragm (-38%)

Cardioprotection in CMH Hamsters

Placebo

GMRD moderate

GMRD severe

GLP Toxicology studies

Rimeporide Clinical trial status update

A complete safety & PK in healthy adults and congestive heart failure patients Ongoing Clinical development in patients with DMD (6 to 14 y)

2015 2016 2017 2021

Acknowledgements to patients & families , investigators and staff & patients organisations

In cohort 1: 5 ambulant DMD patients aged 6 to 10 y, enrolled in

France, received oral rimeporide for 4 W No treatment-related adverse events, no significant

modification of safety lab & tolerability issues In cohort 2: 5 patients (6-10 y) in Italy, Spain & UK

completed, safety analysis ongoing

PK: Visual Predictive Check shows that the adult model,

incorporating allometric scaling, adequately reflects the PK in children

Clinical pharmacology studies in over 166 healthy and CHF patients:

‒ Single oral and IV administration‒ Multiple oral doses administration ‒ Food interaction ‒ Drug Drug Interaction with Digoxin‒ Safety, tolerability, PK in CHF patients with renal insufficiency

Adverse events were mild in intensity with no clear dose dependency

‒ headache (15%), dizziness ( 5%), chest discomfort (10%), paresthesias, vaso-vagal attacks, local skin reactions. No clinically relevant changes in vital signs, ECGs, Laboratory.

‒ Similar events reported on Placebo and treated patients

PK profile in Adults Predicted PK profile in Children

Primary objective: Determine safety & tolerability of Rimeporide after 4 weeks oral treatment Secondary objective : Evaluate the PK profile of rimeporide in plasma Exploratory objectives: NMRI indices (T2, Muscle Mass, FF) & Serum Biomarkers

0

500

1000

1500

2000

2500

3000

0 6 12 18 24 30 36 42 48 54 60 66 72

time [h]

EM

D 8

7 5

80

pla

sma c

on

cen

trati

on

.

[n

g/m

L]

.

mean 50 mg day1

mean 100mg day1

mean 200 mg day1

mean 50 mg day10

mean 100 mg day10

mean 200 mg day10

0 5 10 15 20

050

010

0015

0020

00

Temps (h)

Rim

epor

ide

(µg/

L)

AGE=6yr

AGE=12yr

Extensive Toxicology package did show that Rimeporide was safe

Repeated oral toxicity studies up to 26-week in rats and up to 39-week in dogs showed the main target organ in both species to be fundicparietal cells (reversible mild parietal cells alteration)

No mutagenic activity nor teratogenic potential were shown No impairment of male and female fertility No skin sensitising properties No effect on cardiovascular hemodynamic parameters, no evidence of

risk for any pro-arrhythmic effects No relevant effects on the central and peripheral nervous system, nor

on the gastro-intestinal, renal and respiratory system

Rimeporide

[Na+]i serves as a co-regulator of Ca2+ influx through the NCX and NHE-1, (Burr et al, 2015)

[Na+]ioverload was observed in DMD patients (Weber at al, 2012), is responsible for edema and a secondary increase in basal Ca2+ levels leading to myofiber necrosis and muscles degeneration (Burr et al, 2015)

NHE-1 inhibitors reduce [Na+]i, & indirectly [Ca2+]i (Iwata et al, 2007; Dorchies et al, 2015), reduce myocardial necrosis (Chahine et al, 2005), myofiber fibrosis (Nagaraju et al, 2015), inflammation (Yang et al 2013) and prevent early death (Bkaily et al, 2015) in CM hamsters .

Rimeporide -

Background of NHE-1 inhibition in DMD

Translational GRMD Dogs studies

Cohort 1 PK results

Decreased heart necrosis, thrombosis, myolysis

20 animals (8 severe & 12 moderate) Blinded treatment with Rimeporide at

20 mg/kg/d or placebo started from 2 to 12 months

Objectives: Contribute to the design of the

clinical study by guiding dose selection, identifying novel non invasive biomarkers as well as cardiac outcome measures.

GMRD moderate

Simulated values in ChildrenObserved values in children