Management of retinitis pigmentosa by Wharton's jelly-derived mesenchymal stem cells: prospective analysis of 1-year results

Emin Özmert, Umut Arslan, Emin Özmert, Umut Arslan

Abstract

Purpose: The aim of the study was to investigate annual structural and functional results, and their correlation with inheritance pattern of retinitis pigmentosa (RP) patients who were treated with Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs).

Material and methods: This prospective, sequential, open-label phase-3 clinical study was conducted at Ankara University Faculty of Medicine, Department of Ophthalmology, between April 2019 and May 2020. The study included 34 eyes from 32 retinitis pigmentosa patients of various genotypes who were enrolled in the stem cells clinical trial. The patients were followed for 12 months after the WJ-MSCs transplantation into subtenon space and evaluated with consecutive examinations. Genetic mutations were investigated using a retinitis pigmentosa panel sequencing method consisting of 90 genes. All patients underwent a complete routine ophthalmic examination with best corrected visual acuity, optical coherence tomography angiography, visual field, and full-field electroretinography. Quantitative data obtained from baseline (T0), 6th month (T1), and 12th month (T2) examinations were compared.

Results: According to timepoints at T0, T1, and T2: The mean outer retinal thickness was 100.3 μm, 119.1 μm, and 118.0 μm, respectively (p = 0.01; T0 < T1, T2). The mean horizontal ellipsoid zone width were 2.65 mm, 2.70 mm, and 2.69 mm respectively (p = 0.01; T0 < T1, T2). The mean best corrected visual acuity (BCVA) were 70.5 letters, 80.6 letters, and 79.9 letters, respectively (p = 0.01; T0 < T1, T2). The mean fundus perimetry deviation index (FPDI) was 8.0%, 11.4%, and 11.6%, respectively (p = 0.01; T0 < T1, T2). The mean full-field flicker ERG parameters at T0, T1, and T2: amplitudes were 2.4 mV, 5.0 mV, and 4.6 mV, respectively (p = 0.01; T0 < T1, T2). Implicit time were 43.3 ms, 37.9 ms, and 38.6 ms, respectively (p = 0.01; T0 > T1, T2). According to inheritance pattern, BCVA, FPDI, ERG amplitude, and implicit time data improved significantly in autosomal dominant (AD) and in autosomal recessive (AR) RP at 1 year follow-up (pAD = 0.01, pAR = 0.01; pAD = pAR > pX-linked). No ocular or systemic adverse events related to the surgical methods and/or WJ-MSCs were observed during the 1 year follow-up period.

Conclusion: Subtenon transplantation of WJ-MSCs was found to be effective and safe in the treatment of RP during the first year, similar to the sixth month's results. In autosomal dominant and autosomal recessive inheritance of RP, regardless of the genetic mutations, subtenon administration of WJ-MSCs can be considered an effective and safe option without any adverse effect for slowing or stopping the disease progression.

Trial registration: ClinicalTrials.gov, NCT04224207 . Registered 8 January 2020.

Keywords: Autosomal dominant; Autosomal recessive; Genotype; Retinitis pigmentosa; Stem cell; Wharton jelly.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
a The phenotypic characterization of Wharton jelly-derived mesenchymal stem cells before cryopreservation. b The phenotypic characterization of Wharton jelly-derived mesenchymal stem cells after cryopreservation. c The quantitative results of gene expression products of Wharton jelly-derived mesenchymal stem cells
Fig. 2
Fig. 2
Improvement in “outer retinal thickness” according to study timepoints (T0, T1, T2) in the eye treated with the Wharton’s jelly-derived mesenchymal stem cell (WJ-MSC) (Table 1, patient 1: right eye—published data: a and b are the first 6-month changes of a patient from our previous study [46]). a Before application, 85.0 μm. b At 6th month, 152.2 μm. c At 12th month, 154.1 μm
Fig. 3
Fig. 3
Improvement in “outer retinal thickness” according to study timepoints (T0, T1, T2) in the eye treated with WJ-MSCs (Table 1, patient 4: left eye—published data: a and b are the first 6-month changes of a patient from our previous study [46]). a Before application, 118.2 μm. b At 6th month, 145.4 μm. c At 12th month, 145.6 μm
Fig. 4
Fig. 4
Improvement in “outer retinal thickness” according to study timepoints (T0, T1, T2) in the eye treated with WJ-MSCs (Table 1, patient 15: left eye). a Before application, 84.8 μm. b At 6th month, 120.0 μm. c At 12th month, 122.8 μm
Fig. 5
Fig. 5
Increase in “horizontal ellipsoid zone width” according to study timepoints (T0, T1, T2) in the eye treated with WJ-MSCs (Table 1, patient 26: left eye). a Before application, 2.11 mm. b At 6th month, 2.17 mm. c At 12th month, 2.22 mm
Fig. 6
Fig. 6
Increase in “vertical ellipsoid zone width” according to study timepoints (T0, T1, T2) in the eye treated with the WJ-MSCs (Table 1, patient 24: left eye—the points where the ellipsoid zone intersects the outer limiting membrane are marked as EZ length reference points, as indicated by the arrows). a Before application, 3.21 mm. b At 6th month, 3.34 mm. c At 12th month, 3.35 mm
Fig. 7
Fig. 7
a, b Slowing in the “vertical ellipsoid zone width” loss according to study timepoints (T0, T1, T2) in the eye treated with the WJ-MSCs (Table 1, patient 5: left eye). c, d In the right eye of the same patient without WJ-MSCs treatment, “vertical ellipsoid zone width” decreased rapidly leading to blindness during the 1-year follow-up (X-linked retinitis pigmentosa, GTPase regulator [RPGR] mutation)
Fig. 8
Fig. 8
“Visual field” enlargement according to study timepoints (T0, T1, T2) in the eye treated with WJ-MSCs. Note the change in FPDI and MD values (Table 1, patient 1: right eye—published data: a and b are the first 6-month changes of a patient from our previous study [46]). a Before application, FPDI 5%—MD 28.34. b At 6th month, FPDI 11%—MD 27.72. c At 12th month, FPDI 12%—MD 27.58
Fig. 9
Fig. 9
“Visual field” enlargement according to study timepoints (T0, T1, T2) in the eye treated with WJ-MSCs. Note the change in FPDI and MD values (Table 1, patient 2: left eye—published data: a and b are the first 6-month changes of a patient from our previous study [46]). a Before application, FPDI 6%—MD 28.03. b At 6th month, FPDI 16%—MD 26.61. c At 12th month, FPDI 19%—MD 23.45
Fig. 10
Fig. 10
“Visual field” enlargement according to study timepoints (T0, T1, T2) in the eye treated with WJ-MSCs. Note the change in FPDI (Table 1, patient 6: left eye). a Before application, FPDI 7%. b At 6th month, FPDI 13%. c At 12th month, FPDI 14%

References

    1. Wang AL, Knight DK, Vu TT, Mehta MC. Retinitis pigmentosa: review of current treatment. Int Ophthalmol Clin. 2019;59:263–280.
    1. Zhang Q. Retinitis pigmentosa. Asia-Pac J Ophthalmol. 2016:265–71. 10.1097/apo.0000000000000227.
    1. Hartong DT, Berson EL, Dryja TP. Retinitis pigmentosa. Lancet. 2006:1795–809. 10.1016/s0140-6736(06)69740-7.
    1. Hamel C. Retinitis pigmentosa. Orphanet J Rare Dis. 2006;1:40 4.
    1. Strauss O. The retinal pigment epithelium in visual function. Physiol Rev. 2005;85:845–881.
    1. Cacares PS, Boulan ER. Retinal pigment epithelium polarity in health and blinding diseases. Curr Opin Cell Biol. 2020;62:37–45.
    1. Fuhrmann S, Zou CJ, Levine EM. Retinal pigment epithelium development, plasticity, and tissue homeostasis. Exp Eye Res. 2014;123:141–150.
    1. Dalvi S, Galloway CA, Singh R. Pluripotent stem cells to model degenerative retinal diseases: the RPE perspective. Bharti K.(ed.) Pluripotent stem cells in eye disease therapy, advances in experimental medicine and biology 2019;1186, 10.1007/978-3-030-28471-8_Springer Nature Switzerland.
    1. Ali MU, MSU R, Cao J, Yuan PX. Genetic characterization and disease mechanism of retinitis pigmentosa; current scenario. 3 Biotech. 2017;7(4):251 2.
    1. DiCarlo JE, Mahajan VB, Tsang SH. Gene therapy and genome surgery in the retina. J Clin Invest. 2018;128(6):2177–2188.
    1. Friberg TR. Natural course of retinitis pigmentosa over a three-year interval. Am J Ophthalmol. 1985;100(4):621–622.
    1. Birch DG, Anderson JL, Fish GE. Yearly rates of rod and cone functional loss in retinitis pigmentosa and cone-rod dystrophy. Ophthalmology. 1999;106:258–268.
    1. Liu X, Zhang Y, He Y, Zhao J, Su G. Progress in histopathologic and pathogenetic research in a retinitis pigmentosa model. Histol Histopathol. 2015;30(7):771–779.
    1. Rodríguez-Muñoz A, Aller E, Jaijo T, González-García E, Cabrera-Peset A, et al. Expanding the clinical and molecular heterogeneity of nonsyndromic inherited retinal dystrophies. J Mol Diagn. 2020;22(4):532–543.
    1. Tsang SH, Sharma T. Retinitis pigmentosa (non-syndromic). Atlas Inherit Retin Dis. 2018:125–30. 10.1007/978-3-319-95046-4_25.
    1. Takahashi VKL, Xu CL, Takiuti JT, Apatoff MBL, Duong JK, Mahajan VB, Tsang SH. Comparison of structural progression between ciliopathy and non-ciliopathy associated with autosomal recessive retinitis pigmentosa. Orphanet J Rare Dis. 2019;14:187.
    1. Tsang SH, Aycinena ARP, Sharma T. Ciliopathy: usher syndrome. Atlas Inherit Retin Dis. 2018:167–70. 10.1007/978-3-319-95046-4_32.
    1. Takahashi VKL, Takiuti JT, Carvalho-Jr JRL, Xu CL, Duong JK, Mahajan VB, Tsang SH. Fundus autofluorescence and ellipsoid zone (EZ) line width can be an outcome measurement in RHO-associated autosomal dominant retinitis pigmentosa. Graefes Arch Clin Exp Ophthalmol. 2019;257:725–731.
    1. Tsang SH, Sharma T. Autosomal dominant retinitis pigmentosa. Atlas Inherit Retin Dis. 2018:69–77. 10.1007/978-3-319-95046-4_15.
    1. Cai CX, Locke KG, Ramachandran R, Birch DG, Hood DC. A comparison of progressive loss of the ellipsoid zone (EZ) band in autosomal dominant and x-linked retinitis pigmentosa. Invest Ophthalmol Vis Sci. 2014;23(55):7417–7422.
    1. Sandberg MA, Rosner B, Weigel-DiFranco C, Dryja TP, Berson EL. Disease course of patients with X-linked retinitis pigmentosa due to RPGR gene mutations. Invest Ophthalmol Vis Sci. 2007;48:1298–1304.
    1. Tsang SH, Sharma T. X-linked retinitis pigmentosa. Atlas Inherit Retin Dis. 2018:31–5. 10.1007/978-3-319-95046-4_8.
    1. Diakatou M, Manes G, Bocquet B, Meunier I, Kalatzis V. Genome editing as a treatment for the most prevalent causative genes of autosomal dominant retinitis pigmentosa. Int J Mol Sci. 2019;20(10):2542.
    1. Klassen H. Stem cells in clinical trials for treatment of retinal degeneration. Expert Opin Biol Ther. 2015;16(1):7–14.
    1. Terrell D, Comander J. Current stem-cell approaches for the treatment of inherited retinal degenerations. Semin Ophthalmol. 2019:1–6. 10.1080/08820538.2019.1620808.
    1. Stern JH, Tian Y, Funderburgh J, Pellegrini G, Zhang K, et al. Regenerating eye tissues to preserve and restore vision. Cell Stem Cell. 2018;22(6):834–849.
    1. Leow SN, Luu CD, HairulNizam MH, Mok PL, Ruhaslizan R, Wong HS, et al. Safety and efficacy of human Wharton's jelly-derived mesenchymal stem cells therapy for retinal degeneration. PLoS One. 2015;10(6):e0128973.
    1. Canto-Soler V, Flores-Bellver M, Vergara MN. Stem cell sources and their potential for the treatment of retinal degenerations. Invest Ophthalmol Vis Sci. 2016;57(5):ORSFd1–ORSFd9.
    1. Garg A, Yang J, Lee W, Tsang SH. Stem cell therapies in retinal disorders. Cells. 2017;6(1). doi: 10.3390/cells6010004.
    1. Mohamed EM, Abdelrahman SA, Hussein S, Shalaby SM, Mosaad H, Awad AM. Effect of human umbilical cord blood mesenchymal stem cells administered by intravenous or intravitreal routes on cryo-induced retinal injury. IUBMB Life. 2017;69(3):188–201.
    1. Park SS, Moisseiev E, Bauer G, Anderson JD, Grant MB, et al. Advances in bone marrow stem cell therapy for retinal dysfunction. Prog Retin Eye Res. 2017;56:148–165.
    1. Aladdad AM, Kador KE. Adult stem cells, tools for repairing the retina. Curr Ophthalmol Rep. 2019. 10.1007/s40135-019-00195-z.
    1. Lund RD, Wang S, Lu B. Cells isolated from umbilical cord tissue rescue photoreceptors and visual functions in a rodent model of retinal disease. Stem Cells. 2007;25:602–611.
    1. Rivero JEM, Nicolás FMN, Bernal DG, et al. Human Wharton’s jelly mesenchymal stem cells protect axotomized rat retinal ganglion cells via secretion of antiinflammatory and neurotrophic factors. Sci Rep. 2018;8:16299.
    1. Ruiz FL, Romero CG, Bernal GD, et al. Mesenchymal stromal cell therapy for damaged retinal ganglion cells, is gold all that glitters? Neural Regen Res. 2019;14(11):1851–1857.
    1. Ji S, Lin S, Chen J, Huang X, Wei CC, Li Z, Tang S. Neuroprotection of transplanting human umbilical cord mesenchymal stem cells in a microbead induced ocular hypertension rat model. Curr Eye Res. 2018. 10.1080/02713683.2018.1440604.
    1. Choi SW, Kim JJ, Seo MS, Park SB, Shin TH, et al. Inhibition by miR-410 facilitates direct retinal pigment epithelium differentiation of umbilical cord blood-derived mesenchymal stem cells. J Vet Sci. 2017;18(1):59–65.
    1. Park SS. Cell therapy applications for retinal vascular diseases: diabetic retinopathy and retinal vein occlusion. Invest Ophthalmol Vis Sci. 2016;57:ORSFj1–ORSFj10.
    1. Zhang W, Wang Y, Kong J, Dong M, Duan H, Chen S. Therapeutic efficacy of neural stem cells originating from umbilical cord-derived mesenchymal stem cells in diabetic retinopathy. Sci Rep. 2017;7:408.
    1. Pakuluk AC, Marycz K. A promising tool in retina regeneration: current perspectives and challenges when using mesenchymal progenitor stem cells in veterinary and human ophthalmological applications. Stem Cell Rev Rep. 2017;13:598–602.
    1. Ezquer M, Urzua CA, Montecino S, Leal K, Conget P, Ezquer F. Intravitreal administration of multipotent mesenchymal stromal cells triggers a cytoprotective microenvironment in the retina of diabetic mice. Stem Cell Res Ther. 2016;7:42.
    1. Oner A, Gonen ZB, Sinim N, Cetin M, Ozkul Y. Subretinal adipose tissue-derived mesenchymal stem cell implantation in advanced stage retinitis pigmentosa: a phase I clinical safety study. Stem Cell Res Ther. 2016;7(1). 10.1186/s13287-016-0432-y.
    1. Limoli PG, Limoli C, Vingolo EM, Scalinci SZ, Nebbioso M. Cell surgery and growth factors in dry age-related macular degeneration: visual prognosis and morphological study. Oncotarget. 2016;7(30):46913–46923.
    1. Fiori A, Terlizzi V, Kremer H, Gebauer J, Hammes HP, Harmsen MC, Bieback K. Mesenchymal stromal/stem cells as potential therapy in diabetic retinopathy. Immunobiology. 2018;223(12):729–743.
    1. Bracha P, Moore NA, Ciulla TA. Induced pluripotent stem cell-based therapy for age-related macular degeneration. Expert Opin Biol Ther. 2017;17(9):1113–1126.
    1. Özmert E, Arslan U. Management of retinitis pigmentosa by Wharton's jelly derived mesenchymal stem cells: preliminary clinical results. Stem Cell Res Ther. 2020;11(1):25.
    1. Boya P, Esteban-Martínez L, Serrano-Puebla A, Gómez-Sintes R, Villarejo-Zori B. Autophagy in the eye: development, degeneration, and aging. Prog Retin Eye Res. 2016;55:206–245.
    1. El-Asrag ME, Sergouniotis PI, McKibbin M, Plagnol V, Sheridan E, et al. Biallelic mutations in the autophagy regulator DRAM2 cause retinal dystrophy with early macular involvement. Am J Hum Genet. 2015;96(6):948–954.
    1. Julian JL, Bauer DE, Kong M, Harris MH, Li C, Lindsten T, Thompson CB. Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell. 2005;120(2):237–248.
    1. Lei L, Tzekov R, Li H, McDowell J, Gao G, et al. Inhibition or stimulation of autophagy affects early formation of lipofuscin-like autofluorescence in the retinal pigment epithelium cell. Int J Mol Sci. 2017;18(4):728.
    1. Collins MK, Perkins GR, Rodriguez-Tarduchy G, Nieto MA, López-Rivas A. Growth factors as survival factors: regulation of apoptosis. BioEssays. 1994;16(2):133–138 56.
    1. Marigo V. Programmed cell death in retinal degeneration: targeting apoptosis in photoreceptors as potential therapy for retinal degeneration. Cell Cycle. 2007;6(6):652–655.
    1. Abbaspanah B, Momeni M, Ebrahimi M, Mousavi SH. Advances in perinatal stem cells research: a precious cell source for clinical applications. Regen Med. 2018;13(5):595–610.
    1. Wysocka AM, Kot M, Sułkowski M, Badyra B, Majka M. Molecular and functional verification of Wharton’s jelly mesenchymal stem cells (WJ-MSCs) Pluripotency. Int J Mol Sci. 2019;20:1807.
    1. Bai L, Shao H, Wang H, Zhang Z, Su C, Dong L, Yu B, Chen X, Li X, Zhang X. Effects of mesenchymal stem cell-derived exosomes on experimental autoimmune uveitis. Sci Rep. 2017;7(1):4323.
    1. Rani S, Ryan AE, Griffin MD, Ritter T. Mesenchymal stem cell-derived extracellular vesicles: toward cell-free therapeutic applications. Mol Ther. 2015;23(5):812–823.
    1. Ophelders DR, Wolfs TG, Jellema RK, Zwanenburg A, Andriessen P, et al. Mesenchymal stromal cell-derived extracellular vesicles protect the fetal brain after hypoxia-ischemia. Stem Cells Transl Med. 2016;5(6):754–763.
    1. Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, et al. Minimal information for studies of extracellular vesicles: a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7(1):1535750.
    1. Wang L, Li P, Tian Y. Human umbilical cord mesenchymal stem cells: subpopulations and their difference in cell biology and effects on retinal degeneration in RCS rats. Curr Mol Med. 2017;17:6.
    1. Wu M, Zhang R, Zou Q, Chen Y, Zhou M, et al. Comparison of the biological characteristics of mesenchymal stem cells derived from the human placenta and umbilical cord. Sci Rep. 2018;8:5014.
    1. Karahuseyınoglu S, Çınar Ö, Kılıç E, Kara F, Akay GG, et al. Biology of stem cells in human umbilical cord stroma: in situ and in vitro surveys. Stem Cells. 2007;25:319–331.
    1. Daftarian N, Kiani S, Zahabi A. Regenerative therapy for retinal disorders. J Ophthalmic Vis Res. 2010;5(4):250–264.
    1. Langhe R, Pearson RA. Rebuilding the retina: prospects for Müller glial-mediated self-repair. Curr Eye Res. 2019. 10.1080/02713683.2019.1669665.
    1. Aloe L, Rocco ML, Balzamino BO, Micera A. Nerve growth factor: a focus on neuroscience and therapy. Curr Neuropharmacol. 2015;13:294–303.
    1. Zhang K, Hopkins JJ, Heier JS, Birch DG, Halperin LS, et al. Ciliary neurotrophic factor delivered by encapsulated cell intraocular implants for treatment of geographic atrophy in age-related macular degeneration. Proc Natl Acad Sci U S A. 2011;108(15):6241–6245.
    1. Arslan U, Özmert E, Demirel S, Örnek F, Şermet F. Effects of subtenon-injected autologous platelet-rich plasma on visual functions in eyes with retinitis pigmentosa: preliminary clinical results. Graefes Arch Clin Exp Ophthalmol. 2018;256(5):893–908.
    1. Arslan U, Özmert E. Management of retinitis pigmentosa via platelet-rich plasma or combination with electromagnetic stimulation: retrospective analysis of 1-year results. Adv Ther. 2020;37(5):2390–2412.
    1. Li SK, Hao J. Transscleral passive and iontophoretic transport: theory and analysis. Expert Opin Drug Deliv. 2017;15(3):283–299.
    1. Demetriades AM, Deering T, Liu H, et al. Transscleral delivery of antiangiogenic proteins. J Ocul Pharmacol Ther. 2008;24(1):70–79.
    1. Meng T, Kulkarni V, Simmers R, Brar V, Xu Q. Therapeutic implications of nanomedicine for ocular drug delivery. Drug Discov Today. 2019. 10.1016/j.drudis.2019.05.00.
    1. Mysona BA, Zhao J, Bollinger KE. Role of BDNF/TrkB pathway in the visual system: therapeutic implications for glaucoma. Expert Rev Ophthalmol. 2017;12(1):69–81.
    1. Yoshida N, Ikeda Y, Notomi S, Ishikawa K, Murakami Y, et al. Clinical evidence of sustained chronic inflammatory reaction in retinitis pigmentosa. Ophthalmology. 2013;120:100–105.
    1. Giacalone JC, Andorf JL, Zhang Q, Burnight ER, Ochoa D, et al. Development of a molecularly stable gene therapy vector for the treatment of RPGR-associated X-linked retinitis pigmentosa. Hum Gene Ther. 2019;30(8):967–974.
    1. Miraldi Utz V, Coussa RG, Antaki F, Traboulsi EI. Gene therapy for RPE65-related retinal disease. Ophthalmic Genet. 2018;39(6):671–677.
    1. Munder MC, Midtvedt D, Franzmann T, Nüske E, Otto O, et al. A pH-driven transition of the cytoplasm from a fluid- to a solid-like state promotes entry into dormancy. Elife. 2016;5:e09347.
    1. Wong F, Kwok SY. The survival of cone photoreceptors in retinitis pigmentosa. JAMA Ophthalmol. 2016;134(3):249–250 11.
    1. Koenekoop RK. Why some photoreceptors die,while others remain dormant: lessons from RPE65 and LRAT associated retinal dystrophies. Ophthalmic Genet. 2011;32(2):126–128 9.
    1. Wang W, Lee SJ, Scott PA, Lu X, Emery D, et al. Two-step reactivation of dormant cones in retinitis pigmentosa. Cell Rep. 2016;15(2):372–385.
    1. Sahel JA, Leveillard T, Picaud S, Dalkara D, Marazova K, et al. Functional rescue of cone photoreceptors in retinitis pigmentosa. Grafes Arch Clin Exp Ophthalmol. 2013;251:1669–1677 13.

Source: PubMed

3
Abonnieren