Initial results from a first-in-human gene therapy trial on X-linked retinitis pigmentosa caused by mutations in RPGR

Jasmina Cehajic-Kapetanovic, Kanmin Xue, Cristina Martinez-Fernandez de la Camara, Anika Nanda, Alexandra Davies, Laura J Wood, Anna Paola Salvetti, M Dominik Fischer, James W Aylward, Alun R Barnard, Jasleen K Jolly, Edmond Luo, Brandon J Lujan, Tuyen Ong, Aniz Girach, Graeme C M Black, Ninel Z Gregori, Janet L Davis, Potyra R Rosa, Andrew J Lotery, Byron L Lam, Paulo E Stanga, Robert E MacLaren, Jasmina Cehajic-Kapetanovic, Kanmin Xue, Cristina Martinez-Fernandez de la Camara, Anika Nanda, Alexandra Davies, Laura J Wood, Anna Paola Salvetti, M Dominik Fischer, James W Aylward, Alun R Barnard, Jasleen K Jolly, Edmond Luo, Brandon J Lujan, Tuyen Ong, Aniz Girach, Graeme C M Black, Ninel Z Gregori, Janet L Davis, Potyra R Rosa, Andrew J Lotery, Byron L Lam, Paulo E Stanga, Robert E MacLaren

Abstract

Retinal gene therapy has shown great promise in treating retinitis pigmentosa (RP), a primary photoreceptor degeneration that leads to severe sight loss in young people. In the present study, we report the first-in-human phase 1/2, dose-escalation clinical trial for X-linked RP caused by mutations in the RP GTPase regulator (RPGR) gene in 18 patients over up to 6 months of follow-up (https://ichgcp.net/clinical-trials-registry/NCT03116113" title="See in ClinicalTrials.gov">NCT03116113). The primary outcome of the study was safety, and secondary outcomes included visual acuity, microperimetry and central retinal thickness. Apart from steroid-responsive subretinal inflammation in patients at the higher doses, there were no notable safety concerns after subretinal delivery of an adeno-associated viral vector encoding codon-optimized human RPGR (AAV8-coRPGR), meeting the pre-specified primary endpoint. Visual field improvements beginning at 1 month and maintained to the last point of follow-up were observed in six patients.

Conflict of interest statement

Competing Interest

R.E.M. scientific cofounder of Nightstar Therapeutics Inc. (now owned by Biogen Inc.); R.E.M., G.C.M.B. scientific advisors to the UK National Health Service National Institute for Health and Care Excellence (NICE) in relation to retinal gene therapy; M.D.F., B.L.L., B.J.L., R.E.M. consulting or on advisory board for Biogen Inc.; R.E.M., M.D.F. named inventors on the patent relating to codon-optimised RPGR gene therapy owned by the University of Oxford (US20180273594A1, originally filed 2015-09-10). M.D.F., G.C.M.B., R.E.M. scientific advisory board to Novartis; E.L., T.O., A.G. were employees of the sponsor of the trial, Nightstar Therapeutics (now Biogen Inc). The views expressed are those of the authors and not necessarily those of the National Health Service, the NIHR or the UK Department of Health.

Figures

Fig. 1. Retinal sensitivity following gene therapy…
Fig. 1. Retinal sensitivity following gene therapy for X-linked retinitis pigmentosa in cohort 3 patients.
Mean retinal sensitivity (decibels, dB) and visual field, (represented by sensitivity heat maps) measured by microperimetry, showed progressive improvement in the treated eye from baseline to 6 months post-treatment. The untreated eye showed no change. Visual acuity, measured by ETDRS chart reading (number of letters), remained stable in both eyes. To minimize learning effects, baseline microperimetry was conducted in triplicate over a 2-day period at visit 1 for all subjects. The third reading was validated as an accurate baseline. At follow-up visits, assessments were conducted once for each eye, as per trial protocol.
Fig. 2. Transient regression of retinal function…
Fig. 2. Transient regression of retinal function in the treated eye associated with subretinal inflammation at 5 weeks post high-dose gene therapy.
At week 5 post treatment with AAV8.coRPGR (6x1010 gp), patient C4.1 noticed some regression of vision in the treated eye following improvements over the preceding week. Microperimetry showed regression of retinal sensitivity and visual field (a), which was associated with subretinal lesions (white arrows) on optical coherence tomography (OCT) (b). Subretinal lesions (white arrows) could be seen to be scattered over the treated area of the macula on the OCT cross-sections (c-e), but spared the fovea (f); visual acuity was unaffected following treatment with a course of oral corticosteroids, the inflammation resolved with corresponding gains in retinal sensitivity recorded at 3 months follow-up.
Fig. 3. Outer nuclear layer changes following…
Fig. 3. Outer nuclear layer changes following RPGR gene therapy.
(a) Full manual segmentation of 121-line scans of the OCT at three months of the central macula in the treated eye of patient C4.1 who received the high-dose AAV8.coRPGR vector. This shows increased thickness of the outer nuclear layer (ONL) compared with baseline. The 1, 3 and 6 mm ETDRS macula grid (right column) show mean sectoral ONL thickness changes (μm) and the heat map of ONL thickness (left column). The map shows an increase in retinal thickness (red) in a ring around the fovea of 10-15μm per quadrant, consistent with the OCT images both in terms of the magnitude and location of the retinal thickness change. (b) No change in retinal thickness was observed in the untreated eye. (c) Emergence of novel anatomical structures over areas of previously degenerate macula (red arrows) in the treated eye at 3 months following high-dose (6x1010 gp) RPGR gene therapy. This led to the OCT appearance of ‘double lines’, consisting of the new layer (red arrows) over the RPE layer (black layer). (d) No outer retinal changes were seen in the untreated eye. Scale bars = 200 μm.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/7104347/bin/EMS85431-f004.jpg
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/7104347/bin/EMS85431-f005.jpg
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/7104347/bin/EMS85431-f006.jpg
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/7104347/bin/EMS85431-f007.jpg
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/7104347/bin/EMS85431-f008.jpg
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/7104347/bin/EMS85431-f009.jpg

References

    1. Bennett J. Taking stock of retinal gene therapy: looking back and moving forward. Mol Ther. 2017;25:1076–1094.
    1. Vandenberghe LH. What is next for retinal gene therapy? Cold Spring Harb Perspect Med. 2015;5 a017442.
    1. Boye SE, Boye SL, Lewin AS, Hauswirth WW. A comprehensive review of retinal gene therapy. Mol Ther. 2013;21:509–519.
    1. Sahel JA, Marazova K, Audo I. Clinical characteristics and current therapies for inherited retinal degenerations. Cold Spring Harb Perspect Med. 2014;5 a017111.
    1. Duncan JL, et al. and the Foundation Fighting Blindness Scientific Advisory Board. Inherited Retinal Degenerations: Current Landscape and Knowledge Gaps. Transl Vis Sci Technol. 2018;7(4):6.
    1. DiCarlo JE, Mahajan VB, Tsang SH. Gene therapy and genome surgery in the retina. J Clin Invest. 2018;128:2177–2188.
    1. Talib M, et al. Clinical and genetic characteristics of male patients with RPGR-associated retinal dystrophies: A Long-Term Follow-up Study. Retina. 2018 Mar;8
    1. Fischer MD, et al. Codon-optimized RPGR improves stability and efficacy of AAV8 gene therapy in two mouse models of X-Linked retinitis pigmentosa. Mol Ther. 2017;25:1854–1865.
    1. Baker CK, Flannery JG. Innovative Optogenetic Strategies for Vision Restoration. Front Cell Neurosci. 2018;12:316.
    1. Megaw RD, Soares DC, Wright AF. RPGR: Its role in photoreceptor physiology, human disease, and future therapies. Exp Eye Res. 2015;138:32–41.
    1. Russell S, et al. Efficacy and safety of voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65-mediated inherited retinal dystrophy: a randomised, controlled, open-label, phase 3 trial. Lancet. 2017;390(10097):849–860.
    1. Deng W-T, et al. Stability and safety of an AAV vector for treating RPGR-ORF15X-linked retinitis pigmentosa. Human Gene Therapy. 2015;26(9):593–602.
    1. Wu Z, et al. A long-term efficacy study of gene replacement therapy for RPGR-associated retinal degeneration. Hum Mol Genet. 2015;24(14):3956–3970.
    1. Pawlyk BS, et al. Photoreceptor rescue by an abbreviated human RPGR gene in a murine model of X-linked retinitis pigmentosa. Gene Therapy. 2015;23(2):196–204.
    1. Sun X, et al. Loss of RPGR glutamylation underlies the pathogenic mechanism of retinal dystrophy caused by TTLL5 mutations. Proc Natl Acad Sci U S A. 2016;113(21):E2925–34.
    1. Xue K, Groppe M, Salvetti AP, MacLaren RE. Technique of retinal gene therapy: delivery of viral vector into the subretinal space. Eye (Lond) 2017;31:1308–1316.
    1. Maguire AM, et al. Safety and efficacy of gene transfer for Leber's congenital amaurosis. N Engl J Med. 2008;358(21):2240–8.
    1. Cukras C, et al. Retinal AAV8-RS1 Gene Therapy for X-Linked Retinoschisis: Initial Findings from a Phase I/IIa Trial by Intravitreal Delivery. Mol Ther. 2018;26(9):2282–2294.
    1. Hood DC, Lazow MA, Locke KG, Greenstein VC, Birch DG. The transition zone between healthy and diseased retina in patients with retinitis pigmentosa. Invest Ophthalmol Vis Sci. 2011;52(1):101–8.
    1. Tee JJL, Carroll J, Webster AR, Michaelides M. Quantitative Analysis of Retinal Structure Using Spectral-Domain Optical Coherence Tomography in RPGR-Associated Retinopathy. Am J Ophthalmol. 2017;178:18–26.
    1. Birch DG, et al. Rates of decline in regions of the visual field defined by frequency-domain optical coherence tomography in patients with RPGR-mediated X-linked retinitis pigmentosa. Ophthalmology. 2015;122(4):833–9.
    1. Tang PH, Jauregui R, Tsang SH, Bassuk AG, Mahajan VB. Optical Coherence Tomography Angiography of RPGR-Associated Retinitis Pigmentosa Suggests Foveal Avascular Zone is a Biomarker for Vision Loss. Ophthalmic Surg Lasers Imaging Retina. 2019;50(2):e44–e48.
    1. Duncan JL, et al. High-resolution imaging with adaptive optics in patients with inherited retinal degeneration. Invest Ophthalmol Vis Sci. 2007;48(7):3283–91.
    1. Cideciyan AV, et al. Progression in X-linked Retinitis Pigmentosa Due to ORF15-RPGR Mutations: Assessment of Localized Vision Changes Over 2 Years. Invest Ophthalmol Vis Sci. 2018;59(11):4558–4566.
    1. Bennett J, et al. Safety and durability of effect of contralateral-eye administration of AAV2 gene therapy in patients with childhood-onset blindness caused by RPE65 mutations: a follow-on phase 1 trial. Lancet. 2016;388:661–72.
    1. Storer BE. Design and analysis of phase I clinical trials. Biometrics. 1989;45(3):925–37.
    1. Klein R, Klein BE, Moss SE, DeMets D. Inter-observer variation in refraction and visual acuity measurement using a standardized protocol. Ophthalmology. 1983;90(11):1357–9.
    1. MacLaren RE, et al. Retinal gene therapy in patients with choroideremia: initial findings from a phase 1/2 clinical trial. Lancet. 2014;383(9923):1129–37.
    1. Maguire AM, et al. Safety and efficacy of gene transfer for Leber's congenital amaurosis. N Engl J Med. 2008;358(21):2240–8.

Source: PubMed

3
Sottoscrivi