Gene therapy for Leber's congenital amaurosis is safe and effective through 1.5 years after vector administration

Francesca Simonelli, Albert M Maguire, Francesco Testa, Eric A Pierce, Federico Mingozzi, Jeannette L Bennicelli, Settimio Rossi, Kathleen Marshall, Sandro Banfi, Enrico M Surace, Junwei Sun, T Michael Redmond, Xiaosong Zhu, Kenneth S Shindler, Gui-Shuang Ying, Carmela Ziviello, Carmela Acerra, J Fraser Wright, Jennifer Wellman McDonnell, Katherine A High, Jean Bennett, Alberto Auricchio, Francesca Simonelli, Albert M Maguire, Francesco Testa, Eric A Pierce, Federico Mingozzi, Jeannette L Bennicelli, Settimio Rossi, Kathleen Marshall, Sandro Banfi, Enrico M Surace, Junwei Sun, T Michael Redmond, Xiaosong Zhu, Kenneth S Shindler, Gui-Shuang Ying, Carmela Ziviello, Carmela Acerra, J Fraser Wright, Jennifer Wellman McDonnell, Katherine A High, Jean Bennett, Alberto Auricchio

Abstract

The safety and efficacy of gene therapy for inherited retinal diseases is being tested in humans affected with Leber's congenital amaurosis (LCA), an autosomal recessive blinding disease. Three independent studies have provided evidence that the subretinal administration of adeno-associated viral (AAV) vectors encoding RPE65 in patients affected with LCA2 due to mutations in the RPE65 gene, is safe and, in some cases, results in efficacy. We evaluated the long-term safety and efficacy (global effects on retinal/visual function) resulting from subretinal administration of AAV2-hRPE65v2. Both the safety and the efficacy noted at early timepoints persist through at least 1.5 years after injection in the three LCA2 patients enrolled in the low dose cohort of our trial. A transient rise in neutralizing antibodies to AAV capsid was observed but there was no humoral response to RPE65 protein. The persistence of functional amelioration suggests that AAV-mediated gene transfer to the human retina does not elicit immunological responses which cause significant loss of transduced cells. The persistence of physiologic effect supports the possibility that gene therapy may influence LCA2 disease progression. The safety of the intervention and the stability of the improvement in visual and retinal function in these subjects support the use of AAV-mediated gene augmentation therapy for treatment of inherited retinal diseases.

Figures

Figure 1
Figure 1
Fundus photographs and corresponding optical coherence tomography images through the fovea are shown for all three subjects at baseline, day 30, and 1.5 years after gene delivery. In subject 2, the epiretinal membrane is visible at baseline (arrow). A full-thickness macular hole is apparent on day 30 and 1.5 years. The adjacent retina remains attached.
Figure 2
Figure 2
Representative long-term follow-up pupillometry results in the three subjects. The pupillary light reflexes after dark adaptation are shown as a response to alternating stimulation of the right eye and the left eye. The curves represent the diameter of the pupils; the right pupil is slightly higher than the left pupil in each trace where two pupils are shown. The following pupil traces were displaced vertically to facilitate comparisons: subject 1 day 545, subject 2 baseline, day 365, day 545; subject 3 baseline, subject 3 day 365, subject 3 day 545. The average pupil diameters immediately prior to the first light exposure in the series were as follows: subject 1, 6.4 mm for both pupils at day 175; 5.6 (right), 5.28 (left) at day 446, and 4.9 (right) at day 545; subject 2: 7.1 for both pupils at baseline, 7.8 mm (right) and 7.1 mm (left) pupil at day 60; 4.7 mm for both pupils at day 365 and 4.96 mm (left) pupil at day 545; subject 3: 6.6 mm for both pupils at baseline; 6.3 (right) and 6.1 (left) at day 30; 5.8 (right), 5.4 at day 305 and 4.56 mm at day 545. Alternating stimulation with light of 10.0 lux, 10.0 lux, and 0.04 lux, respectively for subjects 1, 2, and 3. Timepoints (days) with respect to injection are indicated to the right of the traces. Results for test 1 [exposure of eyes to alternating brief (0.2 seconds) flashes of light] are shown for subjects 1 and 2; results for test 2 [exposure of eyes to alternating long (1.0 seconds) flashes of light] are shown for subject 3. Alternating stimuli were presented 2 seconds after the recording was initiated.
Figure 3
Figure 3
Pre- and postinjection nystagmus analysis. (a,b) Frames from pre- and postinjection video recordings in subject 1 with motion paths of the pupils superimposed. Motion of each eye is shown by indicating the location of the center of the pupil over 100 sequential frames (total of 4 seconds) isolated from the video recordings. In b, the motion path is isolated. In c, pre- and postinjection measurements in subject 1 show reduction in interpupillary distance with concomitant changes in the corneal light reflexes (arrowheads). Similar analyses are shown for (d–f) subject 2 and (g–i). Analyses show a significant decrease in binocular amplitude of nystagmus in primary position in subjects 2 and 3 after injection and a smaller decrease in amplitude in subject 1 after injection. There is also a reduction in interpupillary distance in (i) subject 3.
Figure 4
Figure 4
Visual acuity (LogMAR on the left y axis and Snellen value on the right y axis) over time after gene delivery. Baseline is indicated as day 0; gene delivery (no visual acuity measurement) is on day 1.

References

    1. Cremers FP, van den Hurk JA., and , den Hollander AI. Molecular genetics of Leber congenital amaurosis. Hum Mol Genet. 2002;11:1169–1176.
    1. Fulton AB, Hansen RM., and , Mayer DL. Vision in Leber congenital amaurosis. Arch Ophthalmol. 1996;114:698–703.
    1. Aleman TS, Jacobson SG, Chico JD, Scott ML, Cheung AY, Windsor EA, et al. Impairment of the transient pupillary light reflex in Rpe65(−/−) mice and humans with leber congenital amaurosis. Invest Ophthalmol Vis Sci. 2004;45:1259–1271.
    1. Simonelli F, Ziviello C, Testa F, Rossi S, Fazzi E, Bianchi PE, et al. Clinical and molecular genetics of Leber's congenital amaurosis: a multicenter study of Italian patients. Invest Ophthalmol Vis Sci. 2007;48:4284–4290.
    1. den Hollander AI, Roepman R, Koenekoop RK., and , Cremers FP. Leber congenital amaurosis: genes, proteins and disease mechanisms. Prog Retin Eye Res. 2008;27:391–419.
    1. Wang H, den Hollander AI, Moayedi Y, Abulimiti A, Li Y, Collin RW, et al. Mutations in SPATA7 cause Leber congenital amaurosis and juvenile retinitis pigmentosa. Am J Hum Genet. 2009;84:380–387.
    1. Jacobson SG, Aleman TS, Cideciyan AV, Sumaroka A, Schwartz SB, Windsor EA, et al. Identifying photoreceptors in blind eyes caused by RPE65 mutations: Prerequisite for human gene therapy success. Proc Natl Acad Sci USA. 2005;102:6177–6182.
    1. Acland GM, Aguirre GD, Bennett J, Aleman TS, Cideciyan AV, Bennicelli J, et al. Long-term restoration of rod and cone vision by single dose rAAV-mediated gene transfer to the retina in a canine model of childhood blindness. Mol Ther. 2005;12:1072–1082.
    1. Acland GM, Aguirre GD, Ray J, Zhang Q, Aleman TS, Cideciyan AV, et al. Gene therapy restores vision in a canine model of childhood blindness. Nat Genet. 2001;28:92–95.
    1. Bennicelli J, Wright JF, Komaromy A, Jacobs JB, Hauck B, Zelenaia O, et al. Reversal of blindness in animal models of leber congenital amaurosis using optimized AAV2-mediated gene transfer. Mol Ther. 2008;16:458–465.
    1. Bainbridge JW, Smith AJ, Barker SS, Robbie S, Henderson R, Balaggan K, et al. Effect of gene therapy on visual function in Leber's congenital amaurosis. N Engl J Med. 2008;358:2231–2239.
    1. Cideciyan AV, Aleman TS, Boye SL, Schwartz SB, Kaushal S, Roman AJ, et al. Human gene therapy for RPE65 isomerase deficiency activates the retinoid cycle of vision but with slow rod kinetics. Proc Natl Acad Sci USA. 2008;105:15112–15117.
    1. Hauswirth WW, Aleman TS, Kaushal S, Cideciyan AV, Schwartz SB, Wang L, et al. Treatment of leber congenital amaurosis due to RPE65 mutations by ocular subretinal injection of adeno-associated virus gene vector: short-term results of a phase I trial. Hum Gene Ther. 2008;19:979–990.
    1. Maguire AM, Simonelli F, Pierce EA, Pugh EN, Jr, Mingozzi F, Bennicelli J, et al. Safety and efficacy of gene transfer for Leber's congenital amaurosis. N Engl J Med. 2008;358:2240–2248.
    1. Noble KG., and , Carr RE. Stargardt's disease and fundus flavimaculatus. Arch Ophthalmol. 1979;97:1281–1285.
    1. Jacobs JB, Dell'Osso LF, Hertle RW, Acland GM., and , Bennett J. Eye movement recordings as an effectiveness indicator of gene therapy in RPE65-deficient canines: implications for the ocular motor system. Invest Ophthalmol Vis Sci. 2006;47:2865–2875.
    1. Cross SA, Smith JL., and , Norton EW. Periodic alternating nystagmus clearing after vitrectomy. J Clin Neuroophthalmol. 1982;2:5–11.
    1. Yagasaki T, Sato M, Awaya S., and , Nakamura N. Changes in nystagmus after simultaneous surgery for bilateral congenital cataracts. Jpn J Ophthalmol. 1993;37:330–338.
    1. Cideciyan AV, Hauswirth WW, Aleman TS, Kaushal S, Schwartz SB, Boye SL, et al. Human RPE65 gene therapy for Leber congenital amaurosis: persistence of early visual improvements and safety at 1 year. Hum Gene Ther. 2009;20:999–1004.
    1. Cideciyan AV, Hauswirth WW, Aleman TS, Kaushal S, Schwartz SB, Boye SL, et al. Vision 1 year after gene therapy for Leber's congenital amaurosis. N Engl J Med. 2009;361:725–727.
    1. Jiang H, Pierce GF, Ozelo MC, de Paula EV, Vargas JA, Smith P, et al. Evidence of multiyear factor IX expression by AAV-mediated gene transfer to skeletal muscle in an individual with severe hemophilia B. Mol Ther. 2006;14:452–455.
    1. Kaplitt MG, Feigin A, Tang C, Fitzsimons HL, Mattis P, Lawlor PA, et al. Safety and tolerability of gene therapy with an adeno-associated virus (AAV) borne GAD gene for Parkinson's disease: an open label, phase I trial. Lancet. 2007;369:2097–2105.
    1. Manno CS, Pierce GF, Arruda VR, Glader B, Ragni M, Rasko JJ, 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.
    1. Marmor MF, Holder GE, Seeliger MW., and , Yamamoto S.International Society for Clinical Electrophysiology of Vision (2004Standard for clinical electroretinography (2004 update) Doc Ophthalmol 108107–114.
    1. Kawasaki A, Moore P., and , Kardon RH. Variability of the relative afferent pupillary defect. Am J Ophthalmol. 1995;120:622–633.
    1. Ross DF, Fishman GA, Gilbert LD., and , Anderson RJ. Variability of visual field measurements in normal subjects and patients with retinitis pigmentosa. Arch Ophthalmol. 1984;102:1004–1010.

Source: PubMed

3
S'abonner