Abstract
Background:
Adeno-associated virus (AAV) vectors are a promising platform for in vivo transfer of transgenes designed to treat diseases. Pre-existing humoral immunity to these vectors can potentially impact the safety and efficacy of gene therapies. Consequently, individuals with pre-existing antibodies to the specific AAV serotypes used may be excluded from clinical trials and treatments. Recombinant AAV serotype rh74 (rAAVrh74), a vector originally isolated from rhesus monkeys and potentially less immunogenic than other serotypes isolated from humans (e.g. AAV2, AAV5, and AAV9), efficiently transduces muscle and is being investigated for use in gene therapy for Duchenne muscular dystrophy (DMD).
Objective:
To evaluate prevalence of total binding antibodies (neutralizing and non-neutralizing) against rAAVrh74 in patients with DMD.
Methods:
Eligible individuals (N = 107) were ⩾ 4 to < 18 years old with genetically confirmed DMD and were excluded from the study if they lived with a person who had known exposure to rAAVrh74 or other gene transfer therapy, or if they received prior treatment with gene transfer therapy. A single blood sample was obtained from each participant, and anti-rAAVrh74 total binding antibodies were measured by enzyme-linked immunosorbent assay. Total binding antibody level < 1:400 was defined as not elevated or seronegative. Primary endpoint was the percentage of subjects with elevated total antibody titers to rAAVrh74.
Results:
A large preponderance (86.1%) of patients with DMD in this data set was seronegative for anti-rAAVrh74 total binding antibodies. These patients would potentially meet the antibody status eligibility criterion for entry into rAAVrh74-based gene therapy clinical trials.
Conclusion:
Measuring total binding antibodies is a more comprehensive approach to assess pre-existing immune response versus measuring neutralizing antibodies alone. The low seroprevalence of total binding antibodies against rAAVrh74 shown here supports the broad applicability of rAAVrh74-based gene transfer therapy for patients with DMD and potentially other neuromuscular diseases.
Keywords: AAVrh74, antibodies, Duchenne muscular dystrophy, gene transfer therapy, immunity, rAAVrh74
Introduction
Adeno-associated virus (AAV) is a leading gene delivery platform for the treatment of monogenic diseases, with > 200 ongoing or completed clinical trials.1,2 Attributes of AAV include replication deficiency, no known human pathogenicity, and generally weak immunogenicity.3–5 Recombinant AAV vectors used for gene therapy are classified by serotypes that may vary significantly in relative transduction efficiency, tissue tropism, and safety profiles.6–8 Pre-existing humoral immunity to the AAV capsid has the potential to limit therapeutic efficacy and poses a potential safety risk for treatment with AAV-based gene transfer therapies. Specifically, pre-existing AAV antibodies can inhibit transduction9 and promote inflammatory responses via opsonization of AAV particles.10 Furthermore, AAV antibodies have been proposed to stimulate complement activation in clinical trials utilizing AAV9, resulting in severe thrombotic microangiopathic (TMA) events.11–16 Pre-existing antibodies to AAV can either be generated from infection with wild-type AAV or through previous exposure to an AAV-based gene therapy. Antibodies generated against one wild-type AAV can cross-react to multiple AAV serotypes, including recombinant AAV vectors used in gene transfer therapies. Thus, prior to treatment, patients should be screened for pre-existing antibodies specific to the vector capsid being utilized in the given gene therapy.
There is currently no standardized assay for measuring pre-existing immunity against AAV vectors. Rather, entry into a gene transfer therapy program requires a specific antibody test, endorsed by the gene therapy sponsor.2,17 Importantly, program-specific assays can differ in the methodologies used for evaluating pre-existing immunity. Specifically, some sponsors will measure only antibodies that prevent AAV-mediated transduction of target host cells, known as neutralizing antibodies (NAbs), whereas other sponsors will choose a more comprehensive approach that measures total binding antibodies (TAbs), which includes non-neutralizing antibodies (non- NAbs) that can still bind to the vector and promote pro-inflammatory responses in addition to NAbs. Of additional importance in terms of the varied approaches that sponsors take in assessing pre-existing immunity is the cut-off threshold for what is considered to be an ‘elevated’ level of antibodies, or seropositivity, which would preclude participation in a gene therapy program. These thresholds must be experimentally determined through both preclinical and clinical studies and will be unique for each gene therapy program.
As previously mentioned, NAbs are a subset of TAbs capable of inhibiting transduction and thus reducing the efficacy of gene transfer therapy.10,18 NAbs are typically measured using an in vitro cell-based assay, where the neutralizing titer is defined as the highest dilution that reduces transduction by ⩾ 50%. However, the wide variability of assay parameters yields markedly different NAb titers across clinical studies.2 In contrast, TAbs, which include NAbs and non-NAbs, are commonly detected by enzyme-linked immunosorbent assay (ELISA).2 TAb assays measure all antibodies that bind to a specific antigen, irrespective of epitope or neutralizing ability, thus facilitating a comprehensive evaluation of potential impact on safety and efficacy.2,19 TAb assays quantify all antibodies, both NAbs and non-NAbs, that bind to AAV, regardless of their ability to directly block transduction. It is important to screen for non-NAbs because they can have a strong influence on the safety of AAV-based gene therapies by inducing antibody-mediated immune complex formation; opsonization by phagocytic cells, which subsequently promote pro-inflammatory responses; and activation of the classical complement cascade. In addition, high titers of non-NAbs have been shown to reduce the efficacy of gene therapies by promoting premature vector clearance and indirectly influencing the transduction of target tissue.16,20,21
Prevalence of pre-existing antibodies against AAV is dependent on multiple factors, including the serotype, type of antibodies assessed, method of assessment, and geographic location and age range of the population evaluated.17,22 rAAVrh74, which was first identified in rhesus macaques,23 has demonstrated widespread transgene delivery to muscle – including skeletal, diaphragm, and cardiac – in animal models following intravenous administration.5,24,25 Moreover, because rAAVrh74 was isolated from monkeys, it may be associated with less pre-existing immunity compared with AAV serotypes isolated from humans.23,26 These features of rAAVrh74 have made it an attractive platform for development of gene transfer therapies targeting neuromuscular diseases. Indeed, one study that examined seroprevalence of AAV antibodies in various populations, including patients with DMD, found rAAVrh74 to be among the serotypes with the lowest rates of pre-existing immunity.5,23 Several rAAVrh74-based gene transfer therapies are under investigation, including delandistrogene moxeparvovec (SRP-9001) for gene transfer in patients with DMD [phase I (NCT04626674), phase I/II (NCT03375164), phase II (NCT03769116), and phase III (NCT05096221)] and SRP-9003 for the gene transfer of β-sarcoglycan into patients with limb-girdle muscular dystrophy type 2E/R4 [phase I/II (NCT03652259)].
Here, we report the total anti-rAAVrh74 antibody seroprevalence among 101 individuals with DMD. A total antibody level of < 1:400 was defined as seronegative (not elevated) and ⩾ 1:400 was defined as seropositive (elevated). This < 1:400 total antibody threshold was determined based on preclinical nonhuman primate studies20 that found no inhibition of transduction nor safety events caused by antibodies at or below that level; these findings were subsequently clinically validated in human trials.26
Methods
Study design
This study was conducted in the United States through a centralized virtual research coordination center and Principal Investigator, in accordance with the International Conference on Harmonisation Guidelines for Good Clinical Practice and the Declaration of Helsinki. The protocol was approved by a central institutional review board. Eligible patients were males, aged ⩾ 4 to < 18 years at the time of screening, with a definitive diagnosis of DMD based on clinical findings and confirmatory genetic testing. Key exclusion criteria were prior treatment with gene transfer therapy or living with a person who had previous known exposure to rAAVrh74 or other gene transfer therapies. Electronic consent and medical release from the eligible patients’ treating physicians were signed by the patients’ parents/legal guardians. Patients were provided with the choice of receiving their one-time blood draws either at a phlebotomy center or at home, via a home health provider. Blood was collected within 2 weeks of enrollment (serum separator tubes for rAAVrh74 ELISA -). Blood was centrifuged; serum was collected, shipped on cold packs at 2°C–8°C, and then stored at –80°C until analysis. The primary endpoint was the percentage of seropositive patients, which was defined as having elevated anti-rAAVrh74 total antibody titers ⩾ 1:400.
Statistical analysis
The sample size was calculated to obtain 8% precision around the overall seroprevalence estimate. With an estimated sample size of 100 patients, the margin of error [half width of the 95% confidence interval (CI)] would be < 8% for a rate of seroprevalence < 20. Demographic characteristics, including age (years), race, and ethnicity, were summarized for the full analysis set, defined as all patients who were enrolled in the study and had at least one sample evaluated with a valid result for immunogenicity. Analysis of seropositivity was performed on the full analysis set. Clopper–Pearson (exact) method was used to compute the CIs.27
Results
In total, 107 patients with a definitive diagnosis of DMD were enrolled, of whom 101 completed the study (full analysis set) (Figure S1 in the Supplemental material). A summary of patient demographics and baseline characteristics is presented in Table 1. The mean (standard deviation) age of the evaluated patients was 9.1 (3.5) years. Most patients were ⩾ 8 to < 18 years old (65.3%) and White (78.2%). The study was not powered for ethnicity-related analysis. Eighty-one percent of patients selected the home phlebotomy option.
Table 1.
Parameter, years, mean (SD) | Total (N = 101) |
---|---|
Age, years | 9.1 (3.5) |
Years since first motor symptom of DMD to study enrollmenta,b | 6.2 (3.6) |
Years since diagnosis of DMD to study enrollmentb,c | 5.5 (3.5) |
Years since the confirmatory genetic testing to study enrollmentb | 5.3 (3.5) |
Parameter, n (%) | |
Male | 101 (100) |
Race | |
White | 79 (78.2) |
Other | 22 (21.8) |
DMD, Duchenne muscular dystrophy; SD, standard deviation.
n = 94.
Years are calculated as (date of informed consent minus date of the first motor symptom of DMD/DMD diagnosis/the confirmatory genetic testing for DMD)/365.25.
n = 99.
Results from the ELISA showed that a large preponderance of patients [87/101 (86.1%); 95% CI, 77.8–92.2] did not have elevated anti-rAAVrh74 antibody titers (⩾ 1:400) (Figure 1). In the 14 patients with elevated anti-rAAVrh74 antibodies, titers ranged from 1:400 to 1:3200 [1:400: 1.0% (1/101); 1:800: 4.0% (4/101); 1:1600: 5.9% (6/101); and 1:3200: 3.0% (3/101)].
Discussion
Gene therapy has revolutionized the treatment of previously untreatable diseases. There is pressing need for the treatment of devastating, rare, progressive diseases, such as DMD, for which AAV-based gene transfer therapies could significantly impact large segments of the patient population. Currently, some patients are ineligible for AAV-based gene transfer therapies due to pre-existing antibodies to the AAV vector that may affect the safety and efficacy of treatment. In this study, patient samples from a cohort of 101 patients with DMD from across the United States were sent to a single-center laboratory and assessed for seroprevalence of total anti-rAAVrh74 antibodies, where seropositivity, or elevated antibody status, was defined as a titer ⩾ 1:400. This definition for seropositivity was based on preclinical nonhuman primate studies that established a threshold for impact on transduction efficiency. An overwhelming majority of patients were seronegative, suggesting the potential broad applicability of rAAVrh74-based precision genetic therapies for this population.
The innovative virtual design of this study capitalized upon a centralized virtual platform to identify, screen, and receive consent from patients. Moreover, patients were allowed to choose a preferred setting for blood collection (i.e. home versus center-based), thus lowering the overall study burden on healthcare providers, caregivers, and patients. Unforeseen conditions spurred by the COVID-19 pandemic lent further credence and support to the virtual design model.
This seroprevalence study focused on patients with DMD because the rAAVrh74 vector is currently being evaluated in clinical trials of delandistrogene moxeparvovec (SRP-9001), an rAAVrh74-based investigational gene therapy for DMD. Seropositivity for rAAVrh74 is an excluding criterion for clinical trials of rAAVrh74-based therapies. To date, no observed serious adverse events in clinical trials of rAAVrh74-based gene therapies have been attributed to antibody-mediated inflammation or complement activation.26,28 Furthermore, no antibody-mediated inhibition of transduction has been observed in these studies to date. The clinical trials (NCT04626674, NCT03375164, NCT03769116, and NCT03652259) utilize an anti-rAAVrh74 antibody screening test and cut-off definition for elevated TAbs comparable with the test for seropositivity described in this report. The absence of adverse events attributed to anti-rAAVrh74 antibodies in these trials suggests that comprehensive antibody screening is appropriate for determining one aspect of patient eligibility for rAAVrh74 gene therapy.
Ongoing DMD gene therapy clinical development programs in the United States use different mini-/micro-dystrophin transgene constructs, muscle-specific promoters, and AAV serotypes, including rAAVrh74 and AAV9.29 Each gene therapy program requires a unique and specific assay, designed to measure pre-existing antibodies to a particular AAV vector, with threshold values that have ideally been empirically determined and validated through preclinical and clinical studies specific to the vector platform used.
This study based seropositivity on a titer of total binding anti-rAAVrh74 antibodies, or TAbs, which includes NAbs and non-NAbs. Measurement of TAbs, as opposed to simply measuring NAbs, is a more comprehensive approach designed to reduce the risk of adverse events from pre-existing immunity.30,31 Other benefits to TAb assays include their ease of use, their fast turnaround time, and their simplicity to establish and validate compared with complex cell-based NAb assays.30
Limitations of this study include that only United States patients were represented and the use of steroids, which was not assessed. While corticosteroid use has not been shown to impact seroprevalence in other studies of patients with DMD,32 their effect on anti-rAAVrh74 antibody titer was not specifically evaluated in this study; thus, their impact on seroprevalence cannot be ascertained.
In conclusion, results from this study suggest that most patients with DMD in the United States are seronegative (86.1%) for pre-existing antibodies to rAAVrh74. This supports the broad applicability of rAAVrh74-based gene therapy, and those seronegative patients meeting other trial eligibility criteria would be eligible for entry into rAAVrh74-based clinical trials. Finally, because TAb assays offer a more comprehensive approach, this method of patient pre-screening may improve the safety and efficacy of AAV-based gene therapies.
Supplemental Material
Supplemental material, sj-docx-1-tan-10.1177_17562864221149781 for Evaluation of rAAVrh74 gene therapy vector seroprevalence by measurement of total binding antibodies in patients with Duchenne muscular dystrophy by Natalie L. Goedeker, Sachi D. Dharia, Danielle A. Griffin, Jesantha Coy, Todd Truesdale, Rajan Parikh, Kasen Whitehouse, Sourav Santra, Damon R. Asher and Craig M. Zaidman in Therapeutic Advances in Neurological Disorders
Acknowledgments
PPMD provided support in helping to enroll patients and offered optional counseling to patients and caregivers. Audrey Vandervelde of Sarepta Therapeutics, Inc., reviewed the manuscript for scientific accuracy. Medical writing support for this manuscript was provided by Paraskevi Briassouli, PhD, of Eloquent Scientific Solutions, and was funded by Sarepta Therapeutics, Inc.
Footnotes
ORCID iD: Natalie L. Goedeker https://orcid.org/0000-0003-3071-1703
Supplemental material: Supplemental material for this article is available online.
Contributor Information
Natalie L. Goedeker, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8111, St. Louis, MO 63110, USA.
Sachi D. Dharia, Sarepta Therapeutics, Inc., Cambridge, MA, USA
Danielle A. Griffin, Sarepta Therapeutics, Inc., Cambridge, MA, USA
Jesantha Coy, Sarepta Therapeutics, Inc., Cambridge, MA, USA.
Todd Truesdale, Sarepta Therapeutics, Inc., Cambridge, MA, USA.
Rajan Parikh, Sarepta Therapeutics, Inc., Cambridge, MA, USA.
Kasen Whitehouse, Sarepta Therapeutics, Inc., Cambridge, MA, USA.
Sourav Santra, Sarepta Therapeutics, Inc., Cambridge, MA, USA.
Damon R. Asher, Sarepta Therapeutics, Inc., Cambridge, MA, USA
Craig M. Zaidman, Washington University School of Medicine, St. Louis, MO, USA
Declarations
Ethics approval and consent to participate: This study was conducted in the United States through a centralized virtual research coordination center and Principal Investigator, in accordance with the International Conference on Harmonisation Guidelines for Good Clinical Practice and the Declaration of Helsinki. The protocol was approved by a central institutional review board (Advarra; Pro00047571). Electronic consent and medical release from the eligible patients’ treating physicians were signed by the patients’ parents/legal guardians.
Consent for publication: Not applicable.
Author contributions: Natalie L. Goedeker: Data curation; Investigation; Project administration; Resources; Supervision; Writing – review & editing.
Sachi D. Dharia: Formal analysis; Methodology; Writing – review & editing.
Danielle A. Griffin: Formal analysis; Methodology; Writing – review & editing.
Jesantha Coy: Formal analysis; Methodology; Writing – review & editing.
Todd Truesdale: Formal analysis; Methodology; Writing – review & editing.
Rajan Parikh: Formal analysis; Methodology; Writing – review & editing.
Kasen Whitehouse: Formal analysis; Methodology; Writing – review & editing.
Sourav Santra: Formal analysis; Methodology; Writing – review & editing.
Damon R. Asher: Formal analysis; Methodology; Writing – review & editing.
Craig M. Zaidman: Investigation; Writing – review & editing.
Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This study was sponsored by Sarepta Therapeutics, Inc. Funding for the open access charge was provided by Sarepta Therapeutics, Inc.
The authors declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: N.L.G. received consulting fees from Novartis Gene Therapies. D.A., D.G., J.C., K.W., R.P., S.D., S.S., and T.T. are employees of Sarepta Therapeutics, Inc., and may own stock/options in the company. C.M.Z. received research, travel, and speaker fees from, and serves on an advisory board for Biogen, receives research support from Novartis, and was a paid consultant for Optum.
Availability of data and materials: Qualified researchers may request access to the data that support the findings of this study from Sarepta Therapeutics, Inc., by contacting medinfo@sarepta.com.
References
- 1. Kuzmin DA, Shutova MV, Johnston NR, et al. The clinical landscape for AAV gene therapies. Nat Rev Drug Discov 2021; 20: 173–174. [DOI] [PubMed] [Google Scholar]
- 2. Weber T. Anti-AAV antibodies in AAV gene therapy: current challenges and possible solutions. Front Immunol 2021; 12: 658399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Bulaklak K, Xiao X. Therapeutic advances in musculoskeletal AAV targeting approaches. Curr Opin Pharmacol 2017; 34: 56–63. [DOI] [PubMed] [Google Scholar]
- 4. Naso MF, Tomkowicz B, Perry WL, et al. Adeno-associated virus (AAV) as a vector for gene therapy. Biodrugs 2017; 31: 317–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Zygmunt DA, Crowe KE, Flanigan KM, et al. Comparison of serum rAAV serotype-specific antibodies in patients with Duchenne muscular dystrophy, Becker muscular dystrophy, inclusion body myositis, or GNE myopathy. Hum Gene Ther 2017; 28: 737–746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Gao G, Vandenberghe LH, Alvira MR, et al. Clades of adeno-associated viruses are widely disseminated in human tissues. J Virol 2004; 78: 6381–6388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Lau CH, Suh Y. In vivo genome editing in animals using AAV-CRISPR system: applications to translational research of human disease. F1000Res 2017; 6: 2153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Pillay S, Zou W, Cheng F, et al. Adeno-associated virus (AAV) serotypes have distinctive interactions with domains of the cellular AAV receptor. J Virol 2017; 91: e00391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Manno CS, Pierce GF, Arruda VR, et al. Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat Med 2006; 12: 342–347. [DOI] [PubMed] [Google Scholar]
- 10. Calcedo R, Wilson JM. Humoral immune response to AAV. Front Immunol 2013; 4: 341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Chand DH. Clinical findings of thrombotic microangiopathy (TMA). Presented at: cellular, tissue, and gene therapies advisory committee meeting, 2–3 September 2021; Virtual. [Google Scholar]
- 12. Chand DH, Zaidman C, Arya K, et al. Thrombotic microangiopathy following onasemnogene abeparvovec for spinal muscular atrophy: a case series. J Pediatr 2021; 231: 265–268. [DOI] [PubMed] [Google Scholar]
- 13. Hamilton BA, Wright JF. Challenges posed by immune responses to AAV vectors: addressing root causes. Front Immunol 2021; 12: 675897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Pfizer’s new phase 1b results of gene therapy in ambulatory boys with Duchenne muscular dystrophy (DMD) support advancement into pivotal phase 3 study, 15 May 2020, https://www.pfizer.com/news/press-release/press-release-detail/pfizers-new-phase-1b-results-gene-therapy-ambulatory-boys
- 15. Solid Biosciences provides SGT-001 program update, 12 November 2019, https://www.solidbio.com/about/media/press-releases/solid-biosciences-provides-sgt-001-program-update
- 16. Zaiss AK, Cotter MJ, White LR, et al. Complement is an essential component of the immune response to adeno-associated virus vectors. J Virol 2008; 82: 2727–2740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Louis Jeune V, Joergensen JA, Hajjar RJ, et al. Pre-existing anti-adeno-associated virus antibodies as a challenge in AAV gene therapy. Hum Gene Ther Methods 2013; 24: 59–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Wang D, Tai PWL, Gao G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov 2019; 18: 358–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Boutin S, Monteilhet V, Veron P, et al. Prevalence of serum IgG and neutralizing factors against adeno-associated virus (AAV) types 1, 2, 5, 6, 8, and 9 in the healthy population: implications for gene therapy using AAV vectors. Hum Gene Ther 2010; 21: 704–712. [DOI] [PubMed] [Google Scholar]
- 20. Chicoine LG, Montgomery CL, Bremer WG, et al. Plasmapheresis eliminates the negative impact of AAV antibodies on microdystrophin gene expression following vascular delivery. Mol Ther 2014; 22: 338–347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Fitzpatrick Z, Leborgne C, Barbon E, et al. Influence of pre-existing anti-capsid neutralizing and binding antibodies on AAV vector transduction. Mol Ther Methods Clin Dev 2018; 9: 119–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Kruzik A, Fetahagic D, Hartlieb B, et al. Prevalence of anti-adeno-associated virus immune responses in international cohorts of healthy donors. Mol Ther Methods Clin Dev 2019; 14: 126–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Asher DR, Thapa K, Dharia SD, et al. Clinical development on the frontier: gene therapy for Duchenne muscular dystrophy. Expert Opin Biol Ther 2020; 20: 263–274. [DOI] [PubMed] [Google Scholar]
- 24. Pozsgai ER, Griffin DA, Heller KN, et al. Systemic AAV-mediated beta-sarcoglycan delivery targeting cardiac and skeletal muscle ameliorates histological and functional deficits in LGMD2E mice. Mol Ther 2017; 25: 855–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Sondergaard PC, Griffin DA, Pozsgai ER, et al. AAV.dysferlin overlap vectors restore function in dysferlinopathy animal models. Ann Clin Transl Neurol 2015; 2: 256–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Mendell JR, Sahenk Z, Lehman K, et al. Assessment of systemic delivery of rAAVrh74.MHCK7.micro-dystrophin in children with Duchenne muscular dystrophy: a nonrandomized controlled trial. JAMA Neurol 2020; 77: 1122–1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Clopper CJ, Pearson ES. The use of confidence or fiducial limits illustrated in the case of the binomial. Biometrika 1934; 26: 404–413. [Google Scholar]
- 28. Mendell JR. Phase 1/2a trial of delandistrogene moxeparvovec (SRP-9001) in patients with Duchenne muscular dystrophy: 3-year safety and functional outcomes. Poster presentation presented at: muscular dystrophy association (MDA) clinical and scientific conference, Dallas, TX, 13–16 March 2022. [Google Scholar]
- 29. Elangkovan N, Dickson G. Gene therapy for Duchenne muscular dystrophy. J Neuromuscul Dis 2021; 8: S303–S316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Falese L, Sandza K, Yates B, et al. Strategy to detect pre-existing immunity to AAV gene therapy. Gene Ther 2017; 24: 768–778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Long BR, Sandza K, Holcomb J, et al. The impact of pre-existing immunity on the non-clinical pharmacodynamics of AAV5-based gene therapy. Mol Ther Methods Clin Dev 2019; 13: 440–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Verma S, Nwosu SN, Razdan R, et al. Seroprevalence of AAV neutralizing antibodies in males with Duchenne muscular dystrophy. Hum Gene Ther. Epub ahead of print 2 November 2022. DOI: 10.1089/hum.2022.081. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplemental material, sj-docx-1-tan-10.1177_17562864221149781 for Evaluation of rAAVrh74 gene therapy vector seroprevalence by measurement of total binding antibodies in patients with Duchenne muscular dystrophy by Natalie L. Goedeker, Sachi D. Dharia, Danielle A. Griffin, Jesantha Coy, Todd Truesdale, Rajan Parikh, Kasen Whitehouse, Sourav Santra, Damon R. Asher and Craig M. Zaidman in Therapeutic Advances in Neurological Disorders