Presented at WMS | 20-24 September 2021

Microdystrophin gene transfer therapy and therapeutic plasma exchange in nonhuman primates

Ellyn L. Peterson,1 Rachael A. Potter,1 Danielle Griffin,1 Sarah Lewis,1 Eric Pozsgai,1 Aaron Meadows,2 and Louise R. Rodino-Klapac1

1Sarepta Therapeutics, Inc., Cambridge, MA, USA; 2Wexner Medical Center, The Ohio State University, Columbus, OH, USA.

Objectives

Part 1: Investigate the impact of

various immunosuppression strategies

on the safety and efficacy of gene

transfer therapy.

Hypothesis: The duration/regimen of steroids may influence gene transfer therapy safety and transduction efficiency.

Part 2: analyze the safety and efficacy of TPE as a potential pretreatment for individuals with preexisting immunity.

  • Hypothesis: Performing TPE before redosing with gene transfer therapy will reduce AAVrh74 antibody titers, allowing for safer administration in seropositive individuals.

CONCLUSIONS

Part 1: investigate the impact of various immunosuppression strategies on the safety and efficacy of gene transfer therapy

  • Anti-AAVrh74total antibody response to AAVrh74 was similar among all NHP cohorts, with no evidence of abnormal immunologic responses.
  • A few NHPs from cohorts 1-4 experienced transient liver enzyme elevations.
    • This is an expected AE with gene therapy treatment.
    • Levels returned to normal in all cohorts.
  • NHPs from cohort 5 (treated with triple immunosuppression regimen) developed hives 2-3 weeks post vector injection.
    • Two of these NHPs started vomiting 15 weeks post vector injection.
    • AAV titers in cohort 5 never decreased, despite continued sirolimus and an additional rituximab dose at 17 weeks.
  • There were no observed differences in transduction or protein expression with the immunosuppressive regimens tested.

Please scan QR code for full study details

Part 2: analyze the safety and efficacy of TPE as a potential pretreatment for individuals with preexisting immunity

  • The TPE procedure was well tolerated, with no abnormal clinical or immunologic observations.
  • Levels of circulating antibodies to AAVrh74 were reduced after 2-3 consecutive rounds of TPE, and the NHPs that underwent TPE were safely redosed.
  • Further studies are needed to evaluate the safety and efficacy of gene therapy dosing with preexisting immunity to AAVrh74.
  • The presented data suggest TPE as a safe and efficacious strategy to consider for lowering AAVrh74 antibodies.
  • NHPs redosed with high titers experienced significant safety issues. When those titers were reduced with TPE, minimal safety issues were observed. These results highlight the importance of reducing antibodies before dosing.

BACKGROUND

RESULTS Part 1: AAVrh74 antibody response

108

Cohort 1

NHP_12

108

Cohorts 2 and 3

Cohort 2

Part 1: safety profile and transduction efficiency (continued)

Immunosuppression effect in antibody response and transduction efficiency

Part 2: total antibody titers against AAVrh74 in NHP prior to TPE and following TPE (before redosing with SRP-9001) (continued)

  • DMD is a rare, X-linked, and fatal neuromuscular disease caused by mutations in the DMD gene that disrupt the production of functional dystrophin protein.1,2
  • Gene transfer therapy using systemic AAV delivery is being extensively investigated for the treatment of monogenic diseases, including DMD.3
  • A significant challenge to gene transfer therapy is preexisting immunity to AAV vectors, which can result in immune-mediated destruction of transduced cells and limit therapeutic efficacy.3,4
  • Clinical development of gene transfer therapy is advancing rapidly; it is therefore imperative to evaluate strategies to optimize safety and efficacy, as well as dose individuals with preexisting antibodies against the vectors used for delivery.3
  • rAAVrh74.MHCK7.micro-dystrophin(SRP-9001) is a novel AAV-based gene therapy for the

treatment of DMD.3,5

ssDNA

ITR

ITRa

Vector

Promoter

Transgene

PolyAb

5'

3' OH

Dilution)

107

NHP_13

Dilution)

107

NHP_01

NHP_02

106

106

NHP_03

105

105

(1:X

104

(1:X

104

Cohort 3

10000

10000

NHP_04

Plasma

1000

Plasma

1000

NHP_05

100

100

NHP_06

10

10

BSL 2wk 4wk 6wk 8wk

12wk

BSL

2wk 4wk 6wk 8wk

12wk

Weeks Post Gene Transfer

Weeks Post Gene Transfer

Dilution)

107

Cohort 4

NHP_07

Dilution)

108

Cohort 5

NHP_10

107

106

NHP_08

106

NHP_11

105

NHP_09

NHP_14

105

(1:X

104

(1:X

104

Plasma

103

Plasma

103

102

102

101

2wk

4wk 6wk 8wk

12wk

101

BSL 2wk4wk 6wk 8wk

12wk

15wk

BSL

Weeks Post Gene Transfer

Weeks Post Gene Transfer

Anti-AAVrh74 total antibody response was similar across cohorts.

  • No abnormal observations aside from one NHP in cohort 2 that did not mount an antibody response.

The antibody response to AAVrh74 in NHPs in cohort 5 was similar to that of NHPs in cohorts 1-4.

The difference in vector genome copies among NHP cohorts 1-5 was not statistically significant at 12 weeks post gene transfer therapy (P>.05).

Part 2: safety and efficacy analysis of TPE as a potential pretreatment for individuals with preexisting immunity

7 NHPs from cohorts 2-4 underwent 2-3 consecutive cycles of TPE, resulting in

reduced levels of circulating antibodies against AAVrh74.

Antibody titer to AAVrh74 in NHPs following redosing with

108

SRP-9001

NHP_01 (cohort 2)

dilution)

107

NHP_02 (cohort 2)

NHP_03 (cohort 2)

105

NHP_05 (cohort 3)

106

NHP_04 (cohort 3)

(1:X

104

NHP_06 (cohort 3)

NHP_07 (cohort 4)

Plasma

103

NHP_08 (cohort 4)

102

NHP_09 (cohort 4)

101

1 wk

2 wk 4 6

8 10 12

1 d

wk

Weeks post redosing

DottedlinerepresentsinclusioncriteriafortotalAAVrh74antibodytiterlevels(thresholdof1:400againstAAVrh74).

The number of TPE cycles that can be performed in NHPs is limited due to the lack of donor blood available.

In humans, multiple rounds of TPE can be administered.

Important determinant of

Important determinant of safety experience

expression levels

Important determinant of functional impact3,7

and transduction

and specificity3,6

efficiency3,6

EXPRESSION

FUNCTION

SAFETY

SRP-9001

MHCK75

Microdystrophin8

AAVrh748

  1. ITRs are required for genome replication and packaging. b PolyA signals the end of the transgene to the cellular machinery that transcribes (ie, copies) it.

STUDY DESIGN

Part 1

Part 2

All cohorts dosed with

rAAVrh74.MHCK7.microdystrophin

Baselinebiopsyb

(SRP-9001) 2x1014 vg/kg titera

Cohort 4 (n=3)

(SRP-9001) 2x1014 vg/kg

necropsyEOL g

Cohort 4 (n=3)

week-12biopsy

Cohort 1 (n=3)

b

Cohort 1 (n=3)

Cohort 2 (n=3)

Cohort 2 (n=3)

Cohorts 2-5c,d,e

TPE

redosed with

Cohort 3 (n=3)

Cohort 3 (n=3)

rAAVrh74.MHCK7.

microdystrophin

Cohort 5 (n=3)

Cohort 5 (n=3)

titera and

immunosuppressionf

Day −14 GT

Day −1 GT

GT

Day 30 PGT Day 60 PGT

GTh

12 weeks PGT

Prednisone treatment: 2 mg/kg/day

Prednisone treatment (2 mg/kg/day) + rituximab (750 mg/m2) + sirolimus (4 mg/m2/day)i

  1. SupercoiledqPCRtitermethod. bBiopsycollectedfromgastrocnemiusmuscle. cOneNHPdidnotundergoTPEduetolackofantibodyresponsetoAAVrh74. dOneNHPdidnotundergoTPEduetopoorvascularaccess. eCohort 5 did not undergo TPE due to incompatibility with previous treatment with rituximab. fAll NHPs received prednisone(2 mg/kg/day)from 1 day before to 30 days after redosingwithSRP-9001. gEOLnecropsy collected from gastrocnemius, heart, and diaphragm. hImmediately post-TPE, the NHPs were disconnected from the apheresis unit and systemically redosed with rAAVrh74.MHCK7.microdystrophin (SRP-9001). i

Siroliumuswascontinued3daysprioroutpasttheseconddose,rituximab14daysprior,7daysprior,anddayofdosing.

Cohorts 1-4: no abnormal antibody response was observed, except in NHP_03, which did not mount a robust antibody response (despite IV infusion).

Cohort 5: triple immunosuppression regimen did not suppress 2 of 3 NHPs; 1 NHP exhibited slight immunosuppression for 6 weeks (data not shown).

Part 1: safety profile and transduction efficiency

Safety profile (serum chemistry and immunology)

AEs in NHPs from cohorts 1-4 included transient elevated ALT and AST liver enzymes.

NHPs from cohort 5 (treated with a triple immunosuppressive regimen) developed hives 2-3 weeks post vector injection.

  • Benadryl was given orally for 5-10 days.
  • After rituximab infusion 15 weeks post vector injection, 2 patients in cohort 5 began vomiting and experienced elevated heart rates. Vomiting subsided and all other parameters returned to normal ranges when the dose was lowered. Primates recovered from anesthesia without further AEs.

- Immediately after TPE, NHPs were successfully redosed with

rAAVrh74.MHCK7.micro-dystrophin.

- Cohort 5 was redosed without TPE.

In 4 NHPs from cohorts 2-4 (NHP_02, NHP_05, NHP_08, and NHP_09), antibody

titers of ≤1:400 were achieved.

Part 2: total antibody titers against AAVrh74 in NHP prior to TPE and following TPE (before redosing with SRP-9001)

NHP

Titer after part

Titer after TPEb

TPE cycles, n

(cohort)

1a

NHP_01 (2)

1:51200

1:800

2.5

NHP_02 (2)

1:6400

1:400

3

NHP_03 (2)

1:50

NAc

NA

NHP_04 (3)

1:12800

1:800

3

NHP_05 (3)

1:25600

1:400

3

NHP_06 (3)

1:25600

NAd

0.5

NHP_07 (4)

1:12800

1:1600

3

NHP_08 (4)

1:12800

1:200

3

NHP_09 (4)

1:12800

1:200

3

  1. 12 weeks post initial gene transfer. bPrior to redose injection of rAAVrh74.MHCK7.microdystrophin. cNHP_03 was redosed without prior TPE due to lack of antibody response to AAVrh74.dNHP_06onlyunderwent0.5cyclesofTPEduetosmallsizeandpoorvascularaccess.

Part 2: safety profile and immune response to AAVrh74 before and after TPE

Safety profile (serum chemistry and immunology)

The TPE procedure was generally well tolerated.

  • There were no abnormal immunologic observations as assessed by IFN-ꝩ SFC levels against AAVrh74 and microdystrophin peptides from peripheral blood mononuclear cells.
  • Redosing following TPE resulted in increased liver enzyme levels (ALT/AST) in the following NHPs: NHP_01 and NHP_02 (cohort 2), NHP_04 (cohort 3), and NHP_08 and NHP_09 (cohort 4).
    • These were resolved with continued daily administration of prednisone.

NHPs from cohort 5 did not receive TPE due to incompatibility with previous treatmenta and had the total antibody titer to AAVrh74 higher than 1:51,200 before redosing

  • NHPs redosed at high antibody titer (cohort 5) experienced the following AEs: increased heart rate and ventilation rate, vomiting, rash near delivery site, paleness of the skin, and shallow breathing.
    • These all resolved after administration of diphenhydramine and dexamethasone.
  1. Cohort5didnotundergoTPEduetoincompatibilitywithpreviousrituximabtreatment;2NHPs(NHP_10,NHP_11)wereredosed.

REFERENCES

1. Hoffman EP, et al. Cell. 1987;51(6):919-928; 2. Koenig M, et al. Cell. 1987;50(3):509-517; 3. Asher DR, et al. Expert Opin Biol Ther. 2020;20(3):263-274; 4. Manno CS, et al. Nat Med. 2006;12(3):342-347; 5. US National Library of Medicine. Accessed July 2021. https://medlineplus.gov/genetics/understanding/therapy/; 6. Zheng C, Baum BJ. Methods Mol Biol. 2008;434:205-219; 7. Chandler RJ, Venditti CP. Transl Sci Rare Dis. 2016;1(1):73-89; 8. Mendell JR, et al. JAMA Neurol. 2020;77(9):1122- 1131.

ABBREVIATIONS

AAV, adeno-associated virus; AAVrh74, adeno-associated virus rhesus isolate serotype 74; AE, adverse event; ALT, alanine aminotransferase; AST, aspartate aminotransferase; DMD, Duchenne muscular dystrophy; EOL, end of life; FDA, Food and Drug Administration; GT, gene transfer; IFN-ꝩ SFC, interferon gamma spot-forming cells; ITR, inverted terminal repeat; IV, intravenous; MHCK, myosin heavy chain kinase; NA, not available; NHP, nonhuman primate; OH, hydroxyl; PGT, post gene therapy; polyA A, polyadenylation; qPCR, quantitative polymerase chain reaction; rAAVrh74, recombinant AAV rhesus isolate serotype 74; ssDNA, single-stranded DNA; TPE, therapeutic plasma exchange; vg/kg, vector genomes per kilogram bodyweight.

ACKNOWLEDGMENTS AND DISCLOSURES

This study was funded by Sarepta Therapeutics, Inc. SRP-9001 is an investigational therapy and has not been reviewed or approved by the FDA.

ELP, RAP, DG, SL, EP, and LRK are employees of Sarepta Therapeutics and may have stock options. LRK is a coinventor of rAAVrh74.MHCK7.micro- dystrophin technology, which is exclusively licensed to Sarepta Therapeutics. AM was an employee of Sarepta Therapeutics at the time of this study. Medical writing and editorial support was provided by Jen Ciarochi, PhD, of MediTech Media, in accordance with Good Publication Practice (GPP3) guidelines (http://www.ismpp.org/gpp3).

Attachments

  • Original document
  • Permalink

Disclaimer

Sarepta Therapeutics Inc. published this content on 09 September 2021 and is solely responsible for the information contained therein. Distributed by Public, unedited and unaltered, on 20 September 2021 12:21:02 UTC.