Abstract
Age-related macular degeneration (AMD) is a prevalent and complex disease leading to severe vision loss. Stem cells offer promising prospects for AMD treatment as they can be differentiated into critical retinal cell types that could replace lost host retinal cells or provide trophic support to promote host retinal cell survival. However, challenges such as immune rejection, concerns regarding tumorigenicity, and genomic integrity must be addressed. Clinical trials with stem cell-derived retinal pigment epithelial cells have shown preliminary safety in treating dry AMD, but improvements in manufacturing and surgical techniques cell delivery are needed. Late-stage AMD poses additional hurdles, possibly requiring multi-layered grafts. Advancements in automation technologies and gene correction strategies show potential to enhance iPSC-based therapies. Stem cell-based treatments offer hope for AMD management, but further research and optimization are essential for successful clinical implementation.
Introduction
Age-related macular degeneration (AMD) is a complex disease characterized by the loss of the choroidal endothelial cells with subsequent degeneration of the retinal pigment epithelium (RPE) and photoreceptors1. While the exact cause of AMD is not fully understood, oxidative stress, chronic inflammation, genetic factors, and mitochondrial dysfunction are believed to play a role2–4.
Current management options for AMD are limited. In dry AMD, there are emerging treatments, such as pegcetacoplan and avacincaptad pegol and a focus on dietary supplementation to prevent disease progression5–7. Wet AMD can be treated with intravitreal injections of anti-vascular endothelial growth factor (VEGF) drugs, which help limit the growth of abnormal blood vessels in the retina8. Newer drugs include faricimab which targets both VEGF and Ang-2. To date, there is no curative treatment9.
Cell-based therapies using stem cells offer a promising approach for treating AMD10. Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) can be differentiated into cells lost in AMD such as RPE cells, choroidal endothelial cells (CECs), and photoreceptor cells11,12. Several protocols have been developed to differentiate iPSCs into photoreceptor precursor cells, RPE cells, and CECs involving the use of growth factors, chemical molecules, and specific signaling pathways13,14.
Challenges in iPSC-based regenerative medicine include immune rejection, concerns regarding tumorigenicity, genomic integrity, cross-contamination, and sterility15–18. Researchers are addressing these challenges by using immunologically matched donor cells, ensuring the absence of undifferentiated pluripotent stem cells, conducting thorough genetic and karyotypic analyses, and implementing rigorous quality control measures13,19.
Early data from clinical trials have demonstrated promising results, but further research is needed to optimize manufacturing processes, delivery methods, and long-term integration of transplanted cells. While iPSC-based therapies show promise for treating AMD, the treatment of late-stage dry AMD remains a significant challenge due to the loss of both RPE and photoreceptors likely requiring the use of multi-layered grafts to restore visual function20.
AMD background
AMD is the leading cause of blindness in people over 55 in developed countries21 that can lead to severe vision loss. It is estimated that the number of AMD patients will increase from 196 million in 2020 to 288 million in 204022. The disease manifests in two primary forms: dry (atrophic) and wet (neovascular) AMD23,24. The early stages of dry AMD involve pigmentary changes and drusen formation within the macula, the central region of the retina. As dry AMD advances, it can result in geographic atrophy (GA), where loss of photoreceptor cells, RPE cells, and CECs occurs with features visible upon fundoscopic examination25,26. Wet AMD, on the other hand, involves the development of abnormal subretinal choroidal vessels referred to as choroidal neovascular membrane (CNVM). The resulting neovascularization can lead to fluid leakage which is toxic to the retina27. Ultimately, both pathways may lead to the development of a scotoma, or blind spot, in the central visual field sparing the periphery visual field28.
The outer retina, which includes photoreceptor cells and RPE cells, and the choriocapillaris, plays a critical role in vision29,30. Photoreceptor cells convert light into electrical signals, while RPE cells support photoreceptors by providing nutrients and recycling visual pigments. Bruch’s membrane (BrM) serves as a filtering for molecules passing between the choriocapillaris and RPE as well as a barrier to abnormal cellular migration31. The choriocapillaris, as part of the choroid layer beneath the BrM, supplies oxygen and nutrients to the outer retina. In AMD, these layers can be detrimentally affected, with wet AMD involving abnormal blood vessel growth from the choriocapillaris and dry AMD leading to atrophy of the retinal layers, further emphasizing the importance of these structures for maintaining visual health32.
AMD is a complex disease which demonstrates a multifactorial inheritance pattern. The most significant risk factor is age, with individuals over 60 being most susceptible, and the risk increases dramatically with advancing age33,34. Ethnicity also plays a role, with Caucasians at higher risk than African Americans, while global prevalence varies among different ethnic groups35. Cigarette smoking is a notable environmental risk, increasing the odds of AMD development by a factor of almost two compared to non-smokers36. Alcohol use, obesity, and western diets also appear to be modifiable risk factors37. Genetics contribute significantly to AMD risk, with approximately 40 genes associated with the disease, including those involved in the complement cascade38–40. Polymorphisms in complement genes and those in the ARMS2/HTRA1 locus are strongly linked to increased AMD risk41. Understanding these risk factors is crucial for developing targeted interventions and preventative measures for AMD.
Stem cell introduction
Since the advent of induced pluripotent stem cells42,43, stem cell technology, particularly induced pluripotent stem cells (iPSCs), has emerged as a powerful tool for investigating disease. Through patient-specific iPSCs derived with known genetic variation, researchers can gain crucial insights into the mechanism of diseases. This holds especially true for studying human tissues which are otherwise often inaccessible for basic research such as the retina. Moreover, generations of patient-specific cell populations further allow researchers to study the effects of genetic variations in vitro and develop potential targeted therapeutic strategies. To accomplish this, directed differentiation methods aid in generating specific cellular populations, and these cells can be genetically edited to assess the causality of identified variants, distinguishing between necessary and sufficient genetic factors in disease development. For example, iPSC technology has proven effective in understanding monogenic inherited retinal degeneration, leading to the discovery of novel gene variants contributing to retinitis pigmentosa and Usher syndrome44–46. Correcting pathogenic variants in iPSCs using gene editing technologies, such as CRISPR-Cas9, has shown promising results, demonstrating the potential for future therapeutic interventions in retinal disease47–51.
Stem cell technology provides a novel avenue for regenerative medicine. Three sources of stem cells exist for treatment of retinal disease: ESCs, iPSCs, and adult stem cells. Both ESCs and iPSCs are pluripotent and therefore able to differentiate into any cell lineage52 whereas adult stem cells have limited differentiation potential within a specific lineage53. While ESCs have traditionally been used for cell replacement therapy, their use has been subject to both ethical and immunologic concerns. iPSCs and adult stem cells are a viable alternative and, like ESCs, allow for the generation of cells to replace damaged tissue. Multiple past and current clinical trials have undertaken these studies with retinal cells sourced from ESCs, iPSCs, and adult stem cells.
Stem cells to retinal organoids
In one groundbreaking study, Sasai and his collaborators developed a protocol for obtaining eyecup-like structures in vitro54. These studies successfully recreated various aspects of retinal development using retinal organoids (ROs), including the formation of multiple neuronal cell types including, bipolar cells, ganglion cells, horizontal cells, amacrine cells, and Muller cells. Subsequent reports expanded on the utility of the organoid system by identifying stages of organoid maturation and confirming the similarity of RO cell types to those found in fetal and adult retinas through single-cell RNA sequencing analysis46,55–57.
Stem cells to RPE
Methods for deriving RPE cells from stem cells, particularly iPSCs, have been developed to harness their potential for cell-based therapies in AMD. These methods aim to generate functional RPE cells for two reasons: studying disease states in vitro and transplantation into the retina with the goal of restoring visual function10,58.
Differentiation protocols involve the use of specific growth factors, chemical molecules, and signaling pathway inhibitors to guide the iPSCs toward an RPE cell fate. Common factors and molecules utilized include fibroblast growth factor-2 (FGF-2), noggin, activin A, insulin-like growth factor-1 (IGF-1), nicotinamide, WNT antagonists (Dkk-1), bone morphogenetic protein (BMP) antagonists, and small chemical compounds like SB431542 and CHIR9902159–64.
Various differentiation methods have been developed, including spontaneous differentiation, and directed differentiation. Spontaneous differentiation involves changing the culture medium to create an environment conducive to RPE development, although it often yields low efficiency65. Directed differentiation methods use specific combinations of factors and molecules to drive iPSCs directly into the RPE lineage, resulting in more efficient and rapid differentiation66. Validation of RPE differentiation involves evaluating the cells through histological assessment, gene expression analysis, immunofluorescence, and functional assays67.
Despite considerable progress, challenges remain in optimizing the differentiation efficiency, scalability, and functionality of iPSC-derived RPE cells. Additionally, ensuring the safety, sterility, and absence of tumorigenic potential in the derived cells is crucial before considering clinical-grade applications16,68,69.
Stem cells to choroidal endothelial cell
Researchers successfully developed a protocol for generating CECs from iPSCs70. This stepwise protocol employed a lentiviral reporter to track endothelial cell differentiation, transitioning from mTESRTM1 to an endothelial cell medium supplemented with BMP-4, activin A, and FGF-2. Further differentiation into CECs was promoted by VEGF and connective tissue growth factor (CTGF). The research also provided two novel methods of isolating iPSC-derived CECs, leveraging the zeocin resistance offered by the reporter lentivirus versus using anti-CD31 MACS beads.
Importantly, the generated iPSC-derived CECs demonstrated positive staining for CA4 and RGCC, choroid-specific markers, indicating successful differentiation into CECs of the choriocapillaris. Additionally, the cells successfully formed 3-dimensional tube networks – an essential characteristic of mature CECs. In a significant step, these cells were also shown to possess the ability to recellularize decellularized human donor choroidal extracellular matrix.
Autologous vs allogenic
The eye possesses partial immune privilege, attributed to factors such as the blood-ocular barrier and tolerogenic microenvironments71–73. However, breaches in this privilege can occur because of several factors, compromising the success of cellular therapies for eye diseases. Both allogeneic and autologous approaches are being explored for treating age-related macular degeneration, but it remains uncertain which method is most effective and how long transplanted cells can survive in the potentially hostile eye environment74. Studies indicate that short-term systemic immunosuppression may contribute to the long-term survival of allogenic cells, while rejection rates are high without immune suppression. In contrast, immune suppression is unlikely to be needed with autologous cells.
Challenges to autologous iPSCs
Most clinical trials to date have used ESCs due to the significant barriers to iPSC-generated RPE cells. The use of patient-derived iPSCs for autologous cell replacement in clinical trials poses challenges due to the lack of a suitable clinical manufacturing process for simultaneous production and differentiation of multiple patient cell lines74. There have been concerns that reprogramming of somatic cells to iPSCs can introduce genetic alterations in oncogenes and tumor suppressor genes75. While efforts are underway to automate iPSC generation and differentiation, current trials will likely require labor-intensive manual processes, limiting patient enrollment76. Patient selection based on clinical data is critical, but predicting the retinal differentiation capacity of iPSC lines before enrollment is difficult. A recent study identified a gene panel that can predict retinal differentiation capacity of iPSC lines, aiding in cell line selection77. Incorporating assays such as this into the validation pipeline could enhance patient selection and facilitate future interventions to improve retinal differentiation capacity.
Autologous iPSCs are increasingly becoming a viable option for treating retinal diseases as they offer potential advantages over allogenic therapies by bypassing immune response issues. However, a major challenge is the need to correct disease-causing genes in patient-derived cells before transplantation to prevent disease reoccurrence. CRISPR-Cas9 technology shows promise for gene correction, but concerns about unintended genetic modifications remain78. As of current clinical trial data, the necessity of genome correction for known high-risk variants in treating AMD with autologous transplantation remains uncertain. Moreover, off-target effects in genome editing raise safety concerns. To ensure the genetic integrity of donor iPSCs during reprogramming and expansion, single-cell genomics prove valuable for monitoring purposes.
To overcome issues with production capacity, development of automated platforms like CompacT SelecT and CellX, capable of automating iPSC generation and differentiation is vital74,76,79. These systems integrate robotics and liquid handling devices to maintain numerous cell lines in parallel. Utilizing image analysis algorithms in these automated systems could potentially allow for monitoring and validation of cells at every stage of development.
AMD clinical trials
Early cell replacement approaches for treating AMD involved relocating cells from a healthy area of the retina to the site of damage using techniques such as ‘retinal translocation’ 80,81. However, this method posed significant health burdens and surgical risks82. To improve outcomes, researchers explored using RPE allografts in the subretinal space83. Initial trials with cultured fetal RPE patches and cell suspensions showed promise, but emphasized the need for intact Bruch’s membrane and correct RPE orientation for stable engraftment, leading to the development of patch-based cell replacement techniques84. Limited availability of suitable fetal tissue and concerns about tumorigenicity from other cell sources, like aRPE-19, were challenges at the time. Ultimately, this issue was resolved with the advent of stem cell technology.
Multiple clinical trials using stem cells are currently under way (NCT01691261, NCT04339764, NCT03167203, NCT02286089, jRCTa050200027, NCT02590692, NCT01674829). Clinical trials to-date have overall demonstrated safety of transplanting stem cell-derived RPE. In a phase 1/2a study, the safety and preliminary efficacy of human embryonic stem cell (hESC)-derived RPE cells on an ultrathin parylene substrate were evaluated85. Of 15 subjects who received the subretinal implant, the surgery was well-tolerated with manageable adverse events and no implant migration. While underpowered for significance, patients treated trended towards visual improvement. Moreover, the study demonstrated the safety of stem cell-derived RPE and provided encouraging preliminary efficacy data for the treatment of geographic atrophy, leading the authors to conclude that these findings support future recruitment of patients in early-stage disease where there is a greater potential to prevent loss of vision. The first autologous iPSC clinical trial used autologous iPSC-RPE cell sheet in subjects with exudative AMD86. iPSCs were derived from skin fibroblasts and were differentiated into RPE cells. When transplanted into immunodeficient mice, no tumorgenicity was observed. The 1-year results of this trial demonstrated safety without any evidence of visual worsening and overall stability of the transplanted sheets. Lastly, At University of Michigan, we have an ongoing first-of-its-kind clinical trial utilizing adult stem cell-derived RPE cells (NCT04627428). While the results are pending, this source of cells provides another potential avenue for treatment.
Surgical considerations
Central to visual function is the three-neuron circuit consisting of the photoreceptor cell, the bipolar cell, and the ganglion cell, all of which are essential for the detection of light. In the pathology of AMD, the end-stage disease is characterized by the loss of photoreceptor cells, disrupting this crucial circuit. Fortunately, various surgical solutions are available. Two primary surgical approaches exist for accessing the required anatomical space: utilizing either a subretinal bleb or a subretinal incision87. In practice, a bleb creates a localized detachment allowing access to this potential space. At the University of Michigan and in discussions with surgeons involved in similar clinic trials (personal communication), there have been observations that large areas of geographic atrophy can impede subretinal bleb formation due to strong adhesive forces of the overlying, atrophic retina and BrM. Cell delivery can be achieved either as a suspension or through a scaffold. When using suspensions, factors such as the gauge of the delivery system must be considered, as they can affect shearing forces and thus cell viability. Scaffolds may contain one or multiple cell types and might necessitate specialized delivery mechanisms and involve large macular incisions to accommodate cell-scaffold transplants which can lead to hemorrhage and proliferative vitreoretinopathy. RPE replacement may suffice for early-stage disease, although one consideration is that choroidal endothelial cell loss precedes RPE loss in AMD. Late-stage disease presents additional challenges due to photoreceptor cell loss, meaning that treatment for this stage may necessitate a layer of photoreceptor precursor cells to restore visual function. Additionally, the connections between bipolar and ganglion cells must be intact.
Multiple technical challenges exist. First, visualization of the potential space between the RPE and neural retina may be technically challenging during surgery which may be further hindered using non-pigmented RPE cellular suspensions. Intraoperative OCT presents a solution to this hurdle. Other risks include submacular hemorrhage which may present a barrier to transplantation viability, especially those interventions involving large cell-scaffold transplants. Epiretinal membranes have been reported as adverse events presumably due to loss of RPE cells from the transplantation site as well as proliferative vitreoretinopathy88. Lastly, when assessing for efficacy, classic visual acuity metrics may not be sensitive enough to capture meaningful improvements and surrogate measurements may be required such as fundus autofluorescence and microperimetry.
Conclusion
Stem cell-based therapies offer a promising approach for treating AMD. Early clinical trial data indicates safety and potential efficacy. However, further research is needed to optimize manufacturing processes, delivery methods, and long-term integration of transplanted cells. The success of these therapies relies on addressing challenges related to immune rejection, tumorigenicity, and surgical considerations. With ongoing advancements in stem cell technology and surgical techniques, stem cell-based therapies have the potential to revolutionize the treatment of AMD and restore visual function for affected individuals.
Funding
During this work, R.C.R. was supported by the National Eye Institute (R01EY030989, P30EY007003), the National Cancer Institute (P30CA046592, to the University of Michigan Rogel Comprehensive Cancer Center), and Research to Prevent Blindness (Departmental Grant to the University of Michigan Kellogg Eye Center and Career Advancement Award), Beatrice and Reymont Paul Foundation, March Hoops to Beat Blindness, the Taubman Institute, the Leonard G. Miller Endowed Professorship and Ophthalmic Research Fund at the Kellogg Eye Center, and the Grossman, Elaine Sandman, Marek and Maria Spatz (endowed fund), Greenspon, Dunn, Avers, Boustikakis, Sweiden, and Terauchi research funds.
Otherwise there are no other funding sources nor conflicts of interest to disclose.
References
- 1.Ayoub T, Patel N. Age-related macular degeneration. J R Soc Med. Feb 2009;102(2):56–61. doi: 10.1258/jrsm.2009.080298 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Datta S, Cano M, Ebrahimi K, Wang L, Handa JT. The impact of oxidative stress and inflammation on RPE degeneration in non-neovascular AMD. Prog Retin Eye Res. Sep 2017;60:201–218. doi: 10.1016/j.preteyeres.2017.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Kaarniranta K, Hyttinen J, Ryhanen T, et al. Mechanisms of protein aggregation in the retinal pigment epithelial cells. Front Biosci (Elite Ed). Jun 1 2010;2(4):1374–84. doi: 10.2741/e198 [DOI] [PubMed] [Google Scholar]
- 4.Kaarniranta K, Uusitalo H, Blasiak J, et al. Mechanisms of mitochondrial dysfunction and their impact on age-related macular degeneration. Prog Retin Eye Res. Nov 2020;79:100858. doi: 10.1016/j.preteyeres.2020.100858 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Liao DS, Metlapally R, Joshi P. Pegcetacoplan treatment for geographic atrophy due to age-related macular degeneration: a plain language summary of the FILLY study. Immunotherapy. Sep 2022;14(13):995–1006. doi: 10.2217/imt-2022-0078 [DOI] [PubMed] [Google Scholar]
- 6.Wykoff CC, Rosenfeld PJ, Waheed NK, et al. Characterizing New-Onset Exudation in the Randomized Phase 2 FILLY Trial of Complement Inhibitor Pegcetacoplan for Geographic Atrophy. Ophthalmology. Sep 2021;128(9):1325–1336. doi: 10.1016/j.ophtha.2021.02.025 [DOI] [PubMed] [Google Scholar]
- 7.Chew EY, Clemons TE, Agrón E, et al. Ten-year follow-up of age-related macular degeneration in the age-related eye disease study: AREDS report no. 36. JAMA Ophthalmol. Mar 2014;132(3):272–7. doi: 10.1001/jamaophthalmol.2013.6636 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ricci F, Bandello F, Navarra P, Staurenghi G, Stumpp M, Zarbin M. Neovascular Age-Related Macular Degeneration: Therapeutic Management and New-Upcoming Approaches. Int J Mol Sci. Nov 3 2020;21(21)doi: 10.3390/ijms21218242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Mori R, Honda S, Gomi F, et al. Efficacy, durability, and safety of faricimab up to every 16 weeks in patients with neovascular age-related macular degeneration: 1-year results from the Japan subgroup of the phase 3 TENAYA trial. Jpn J Ophthalmol. May 2023;67(3):301–310. doi: 10.1007/s10384-023-00985-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Mandai M, Watanabe A, Kurimoto Y, et al. Autologous Induced Stem-Cell-Derived Retinal Cells for Macular Degeneration. N Engl J Med. Mar 16 2017;376(11):1038–1046. doi: 10.1056/NEJMoa1608368 [DOI] [PubMed] [Google Scholar]
- 11.Bharti K, den Hollander AI, Lakkaraju A, et al. Cell culture models to study retinal pigment epithelium-related pathogenesis in age-related macular degeneration. Exp Eye Res. Sep 2022;222:109170. doi: 10.1016/j.exer.2022.109170 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Dalvi S, Galloway CA, Singh R. Pluripotent Stem Cells to Model Degenerative Retinal Diseases: The RPE Perspective. Adv Exp Med Biol. 2019;1186:1–31. doi: 10.1007/978-3-030-28471-8_1 [DOI] [PubMed] [Google Scholar]
- 13.Watari K, Yamasaki S, Tu HY, et al. Self-organization, quality control, and preclinical studies of human iPSC-derived retinal sheets for tissue-transplantation therapy. Commun Biol. Feb 10 2023;6(1):164. doi: 10.1038/s42003-023-04543-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wiley LA, Burnight ER, DeLuca AP, et al. cGMP production of patient-specific iPSCs and photoreceptor precursor cells to treat retinal degenerative blindness. Sci Rep. Jul 29 2016;6:30742. doi: 10.1038/srep30742 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Petrash CC, Palestine AG, Canto-Soler MV. Immunologic Rejection of Transplanted Retinal Pigmented Epithelium: Mechanisms and Strategies for Prevention. Front Immunol. 2021;12:621007. doi: 10.3389/fimmu.2021.621007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kanemura H, Go MJ, Shikamura M, et al. Tumorigenicity studies of induced pluripotent stem cell (iPSC)-derived retinal pigment epithelium (RPE) for the treatment of age-related macular degeneration. PLoS One. 2014;9(1):e85336. doi: 10.1371/journal.pone.0085336 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kashani AH. Stem cell-derived retinal pigment epithelium transplantation in age-related macular degeneration: recent advances and challenges. Curr Opin Ophthalmol. May 1 2022;33(3):211–218. doi: 10.1097/icu.0000000000000838 [DOI] [PubMed] [Google Scholar]
- 18.Yamanaka S. Pluripotent Stem Cell-Based Cell Therapy-Promise and Challenges. Cell Stem Cell. Oct 1 2020;27(4):523–531. doi: 10.1016/j.stem.2020.09.014 [DOI] [PubMed] [Google Scholar]
- 19.Andrews PW, Barbaric I, Benvenisty N, et al. The consequences of recurrent genetic and epigenetic variants in human pluripotent stem cells. Cell Stem Cell. Dec 1 2022;29(12):1624–1636. doi: 10.1016/j.stem.2022.11.006 [DOI] [PubMed] [Google Scholar]
- 20.Tsai Y, Lu B, Bakondi B, et al. Human iPSC-Derived Neural Progenitors Preserve Vision in an AMD-Like Model. Stem Cells. Aug 2015;33(8):2537–49. doi: 10.1002/stem.2032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Deng Y, Qiao L, Du M, et al. Age-related macular degeneration: Epidemiology, genetics, pathophysiology, diagnosis, and targeted therapy. Genes Dis. Jan 2022;9(1):62–79. doi: 10.1016/j.gendis.2021.02.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wong WL, Su X, Li X, et al. Global prevalence of age-related macular degeneration and disease burden projection for 2020 and 2040: a systematic review and meta-analysis. Lancet Glob Health. Feb 2014;2(2):e106–16. doi: 10.1016/s2214-109x(13)70145-1 [DOI] [PubMed] [Google Scholar]
- 23.Pennington KL, DeAngelis MM. Epidemiology of age-related macular degeneration (AMD): associations with cardiovascular disease phenotypes and lipid factors. Eye Vis (Lond). 2016;3:34. doi: 10.1186/s40662-016-0063-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ambati J, Fowler BJ. Mechanisms of age-related macular degeneration. Neuron. Jul 12 2012;75(1):26–39. doi: 10.1016/j.neuron.2012.06.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Curcio CA, Medeiros NE, Millican CL. Photoreceptor loss in age-related macular degeneration. Invest Ophthalmol Vis Sci. Jun 1996;37(7):1236–49. [PubMed] [Google Scholar]
- 26.The Age-Related Eye Disease Study system for classifying age-related macular degeneration from stereoscopic color fundus photographs: the Age-Related Eye Disease Study Report Number 6. Am J Ophthalmol. Nov 2001;132(5):668–81. doi: 10.1016/s0002-9394(01)01218-1 [DOI] [PubMed] [Google Scholar]
- 27.Ferris FL, 3rd, Fine SL, Hyman L. Age-related macular degeneration and blindness due to neovascular maculopathy. Arch Ophthalmol. Nov 1984;102(11):1640–2. doi: 10.1001/archopht.1984.01040031330019 [DOI] [PubMed] [Google Scholar]
- 28.Nita M, Strzałka-Mrozik B, Grzybowski A, Mazurek U, Romaniuk W. Age-related macular degeneration and changes in the extracellular matrix. Med Sci Monit. Jun 18 2014;20:1003–16. doi: 10.12659/msm.889887 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Masland RH. The neuronal organization of the retina. Neuron. Oct 18 2012;76(2):266–80. doi: 10.1016/j.neuron.2012.10.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Lejoyeux R, Benillouche J, Ong J, et al. Choriocapillaris: Fundamentals and advancements. Prog Retin Eye Res. Mar 2022;87:100997. doi: 10.1016/j.preteyeres.2021.100997 [DOI] [PubMed] [Google Scholar]
- 31.Murali A, Krishnakumar S, Subramanian A, Parameswaran S. Bruch’s membrane pathology: A mechanistic perspective. Eur J Ophthalmol. Nov 2020;30(6):1195–1206. doi: 10.1177/1120672120919337 [DOI] [PubMed] [Google Scholar]
- 32.Nowak JZ. Age-related macular degeneration (AMD): pathogenesis and therapy. Pharmacol Rep. May-Jun 2006;58(3):353–63. [PubMed] [Google Scholar]
- 33.Rein DB, Wittenborn JS, Zhang X, Honeycutt AA, Lesesne SB, Saaddine J. Forecasting age-related macular degeneration through the year 2050: the potential impact of new treatments. Arch Ophthalmol. Apr 2009;127(4):533–40. doi: 10.1001/archophthalmol.2009.58 [DOI] [PubMed] [Google Scholar]
- 34.Rein DB, Wittenborn JS, Burke-Conte Z, et al. Prevalence of Age-Related Macular Degeneration in the US in 2019. JAMA Ophthalmol. Dec 1 2022;140(12):1202–1208. doi: 10.1001/jamaophthalmol.2022.4401 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Klein R, Klein BE, Cruickshanks KJ. The prevalence of age-related maculopathy by geographic region and ethnicity. Prog Retin Eye Res. May 1999;18(3):371–89. doi: 10.1016/s1350-9462(98)00025-1 [DOI] [PubMed] [Google Scholar]
- 36.Kuan V, Warwick A, Hingorani A, et al. Association of Smoking, Alcohol Consumption, Blood Pressure, Body Mass Index, and Glycemic Risk Factors With Age-Related Macular Degeneration: A Mendelian Randomization Study. JAMA Ophthalmol. Dec 1 2021;139(12):1299–1306. doi: 10.1001/jamaophthalmol.2021.4601 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Pugazhendhi A, Hubbell M, Jairam P, Ambati B. Neovascular Macular Degeneration: A Review of Etiology, Risk Factors, and Recent Advances in Research and Therapy. Int J Mol Sci. Jan 25 2021;22(3)doi: 10.3390/ijms22031170 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Fritsche LG, Fariss RN, Stambolian D, Abecasis GR, Curcio CA, Swaroop A. Age-related macular degeneration: genetics and biology coming together. Annu Rev Genomics Hum Genet. 2014;15:151–71. doi: 10.1146/annurev-genom-090413-025610 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Maguire MG, Ying GS, Jaffe GJ, et al. Single-Nucleotide Polymorphisms Associated With Age-Related Macular Degeneration and Lesion Phenotypes in the Comparison of Age-Related Macular Degeneration Treatments Trials. JAMA Ophthalmol. Jun 1 2016;134(6):674–81. doi: 10.1001/jamaophthalmol.2016.0669 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Armento A, Ueffing M, Clark SJ. The complement system in age-related macular degeneration. Cell Mol Life Sci. May 2021;78(10):4487–4505. doi: 10.1007/s00018-021-03796-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Deangelis MM, Silveira AC, Carr EA, Kim IK. Genetics of age-related macular degeneration: current concepts, future directions. Semin Ophthalmol. May 2011;26(3):77–93. doi: 10.3109/08820538.2011.577129 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. Aug 25 2006;126(4):663–76. doi: 10.1016/j.cell.2006.07.024 [DOI] [PubMed] [Google Scholar]
- 43.Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. Nov 30 2007;131(5):861–72. doi: 10.1016/j.cell.2007.11.019 [DOI] [PubMed] [Google Scholar]
- 44.Tucker BA, Scheetz TE, Mullins RF, et al. Exome sequencing and analysis of induced pluripotent stem cells identify the cilia-related gene male germ cell-associated kinase (MAK) as a cause of retinitis pigmentosa. Proc Natl Acad Sci U S A. Aug 23 2011;108(34):E569–76. doi: 10.1073/pnas.1108918108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ukaji T, Takahashi-Shibata M, Arai D, et al. Generation and characterization of a human iPSC line (JUFMDOi007-A) from a patient with Usher syndrome due to mutation in USH2A. Stem Cell Res. Jun 2023;69:103100. doi: 10.1016/j.scr.2023.103100 [DOI] [PubMed] [Google Scholar]
- 46.Lane A, Jovanovic K, Shortall C, et al. Modeling and Rescue of RP2 Retinitis Pigmentosa Using iPSC-Derived Retinal Organoids. Stem Cell Reports. Jul 14 2020;15(1):67–79. doi: 10.1016/j.stemcr.2020.05.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Benati D, Patrizi C, Recchia A. Gene editing prospects for treating inherited retinal diseases. J Med Genet. Jul 2020;57(7):437–444. doi: 10.1136/jmedgenet-2019-106473 [DOI] [PubMed] [Google Scholar]
- 48.Wu X, Yan N, Zhang M. Retinal Degeneration: Molecular Mechanisms and Therapeutic Strategies. Curr Med Chem. 2022;29(40):6125–6140. doi: 10.2174/0929867328666211129122908 [DOI] [PubMed] [Google Scholar]
- 49.Ziccardi L, Cordeddu V, Gaddini L, et al. Gene Therapy in Retinal Dystrophies. Int J Mol Sci. Nov 14 2019;20(22)doi: 10.3390/ijms20225722 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Yang TC, Chang CY, Yarmishyn AA, et al. Carboxylated nanodiamond-mediated CRISPR-Cas9 delivery of human retinoschisis mutation into human iPSCs and mouse retina. Acta Biomater. Jan 1 2020;101:484–494. doi: 10.1016/j.actbio.2019.10.037 [DOI] [PubMed] [Google Scholar]
- 51.Burnight ER, Gupta M, Wiley LA, et al. Using CRISPR-Cas9 to Generate Gene-Corrected Autologous iPSCs for the Treatment of Inherited Retinal Degeneration. Mol Ther. Sep 6 2017;25(9):1999–2013. doi: 10.1016/j.ymthe.2017.05.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Halevy T, Urbach A. Comparing ESC and iPSC-Based Models for Human Genetic Disorders. J Clin Med. Oct 24 2014;3(4):1146–62. doi: 10.3390/jcm3041146 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Salero E, Blenkinsop TA, Corneo B, et al. Adult human RPE can be activated into a multipotent stem cell that produces mesenchymal derivatives. Cell Stem Cell. Jan 6 2012;10(1):88–95. doi: 10.1016/j.stem.2011.11.018 [DOI] [PubMed] [Google Scholar]
- 54.Nakano T, Ando S, Takata N, et al. Self-formation of optic cups and storable stratified neural retina from human ESCs. Cell Stem Cell. Jun 14 2012;10(6):771–785. doi: 10.1016/j.stem.2012.05.009 [DOI] [PubMed] [Google Scholar]
- 55.Kruczek K, Gonzalez-Cordero A, Goh D, et al. Differentiation and Transplantation of Embryonic Stem Cell-Derived Cone Photoreceptors into a Mouse Model of End-Stage Retinal Degeneration. Stem Cell Reports. Jun 6 2017;8(6):1659–1674. doi: 10.1016/j.stemcr.2017.04.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Takata N, Eiraku M, Sakakura E. Data describing Rax positive optic-vesicle generation from mouse embryonic stem cells in vitro. Data Brief. Sep 2016;8:465–9. doi: 10.1016/j.dib.2016.05.070 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Sridhar A, Hoshino A, Finkbeiner CR, et al. Single-Cell Transcriptomic Comparison of Human Fetal Retina, hPSC-Derived Retinal Organoids, and Long-Term Retinal Cultures. Cell Rep. Feb 4 2020;30(5):1644–1659.e4. doi: 10.1016/j.celrep.2020.01.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Surendran H, Rathod RJ, Pal R. Generation of Transplantable Retinal Pigmented Epithelial (RPE) Cells for Treatment of Age-Related Macular Degeneration (AMD). Methods Mol Biol. 2019;2045:283–298. doi: 10.1007/7651_2018_140 [DOI] [PubMed] [Google Scholar]
- 59.Idelson M, Alper R, Obolensky A, et al. Directed differentiation of human embryonic stem cells into functional retinal pigment epithelium cells. Cell Stem Cell. Oct 2 2009;5(4):396–408. doi: 10.1016/j.stem.2009.07.002 [DOI] [PubMed] [Google Scholar]
- 60.Zahabi A, Shahbazi E, Ahmadieh H, et al. A new efficient protocol for directed differentiation of retinal pigmented epithelial cells from normal and retinal disease induced pluripotent stem cells. Stem Cells Dev. Aug 10 2012;21(12):2262–72. doi: 10.1089/scd.2011.0599 [DOI] [PubMed] [Google Scholar]
- 61.Westenskow P, Sedillo Z, Barnett A, Friedlander M. Efficient derivation of retinal pigment epithelium cells from stem cells. J Vis Exp. Mar 8 2015;(97)doi: 10.3791/52214 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Al-Ani A, Sunba S, Hafeez B, Toms D, Ungrin M. In Vitro Maturation of Retinal Pigment Epithelium Is Essential for Maintaining High Expression of Key Functional Genes. Int J Mol Sci. Aug 23 2020;21(17)doi: 10.3390/ijms21176066 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Li J, Qiu C, Wei Y, et al. Human Amniotic Epithelial Stem Cell-Derived Retinal Pigment Epithelium Cells Repair Retinal Degeneration. Front Cell Dev Biol. 2021;9:737242. doi: 10.3389/fcell.2021.737242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Hirami Y, Osakada F, Takahashi K, et al. Generation of retinal cells from mouse and human induced pluripotent stem cells. Neurosci Lett. Jul 24 2009;458(3):126–31. doi: 10.1016/j.neulet.2009.04.035 [DOI] [PubMed] [Google Scholar]
- 65.Ferguson LR, Balaiya S, Mynampati BK, Sambhav K, Chalam KV. Deprivation of bFGF Promotes Spontaneous Differentiation of Human Embryonic Stem Cells into Retinal Pigment Epithelial Cells. J Stem Cells. 2015;10(3):159–70. [PubMed] [Google Scholar]
- 66.Buchholz DE, Pennington BO, Croze RH, Hinman CR, Coffey PJ, Clegg DO. Rapid and efficient directed differentiation of human pluripotent stem cells into retinal pigmented epithelium. Stem Cells Transl Med. May 2013;2(5):384–93. doi: 10.5966/sctm.2012-0163 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Khristov V, Maminishkis A, Amaral J, Rising A, Bharti K, Miller S. Validation of iPS Cell-Derived RPE Tissue in Animal Models. Adv Exp Med Biol. 2018;1074:633–640. doi: 10.1007/978-3-319-75402-4_77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Choudhary P, Whiting PJ. A strategy to ensure safety of stem cell-derived retinal pigment epithelium cells. Stem Cell Res Ther. Sep 2 2016;7(1):127. doi: 10.1186/s13287-016-0380-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Nazari H, Zhang L, Zhu D, et al. Stem cell based therapies for age-related macular degeneration: The promises and the challenges. Prog Retin Eye Res. Sep 2015;48:1–39. doi: 10.1016/j.preteyeres.2015.06.004 [DOI] [PubMed] [Google Scholar]
- 70.Mulfaul K, Giacalone JC, Voigt AP, et al. Stepwise differentiation and functional characterization of human induced pluripotent stem cell-derived choroidal endothelial cells. Stem Cell Res Ther. Sep 23 2020;11(1):409. doi: 10.1186/s13287-020-01903-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Streilein JW. Ocular immune privilege: the eye takes a dim but practical view of immunity and inflammation. J Leukoc Biol. Aug 2003;74(2):179–85. doi: 10.1189/jlb.1102574 [DOI] [PubMed] [Google Scholar]
- 72.Streilein JW, Ohta K, Mo JS, Taylor AW. Ocular immune privilege and the impact of intraocular inflammation. DNA Cell Biol. May-Jun 2002;21(5–6):453–9. doi: 10.1089/10445490260099746 [DOI] [PubMed] [Google Scholar]
- 73.Streilein JW. Immunological non-responsiveness and acquisition of tolerance in relation to immune privilege in the eye. Eye (Lond). 1995;9 ( Pt 2):236–40. doi: 10.1038/eye.1995.46 [DOI] [PubMed] [Google Scholar]
- 74.Fortress AM, Miyagishima KJ, Reed AA, et al. Stem cell sources and characterization in the development of cell-based products for treating retinal disease: An NEI Town Hall report. Stem Cell Res Ther. Mar 29 2023;14(1):53. doi: 10.1186/s13287-023-03282-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Barker RA, Carpenter M, Jamieson CHM, et al. Lessons learnt, and still to learn, in first in human stem cell trials. Stem Cell Reports. Aug 8 2023;18(8):1599–1609. doi: 10.1016/j.stemcr.2022.11.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Bohrer LR, Stone NE, Mullin NK, et al. Automating iPSC generation to enable autologous photoreceptor cell replacement therapy. J Transl Med. Feb 28 2023;21(1):161. doi: 10.1186/s12967-023-03966-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Cooke JA, Voigt AP, Collingwood MA, et al. Propensity of Patient-Derived iPSCs for Retinal Differentiation: Implications for Autologous Cell Replacement. Stem Cells Transl Med. Jun 15 2023;12(6):365–378. doi: 10.1093/stcltm/szad028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Giacalone JC, Sharma TP, Burnight ER, et al. CRISPR-Cas9-Based Genome Editing of Human Induced Pluripotent Stem Cells. Curr Protoc Stem Cell Biol. Feb 28 2018;44:5b.7.1–5b.7.22. doi: 10.1002/cpsc.46 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Mullin NK, Voigt AP, Cooke JA, et al. Patient derived stem cells for discovery and validation of novel pathogenic variants in inherited retinal disease. Prog Retin Eye Res. Jul 2021;83:100918. doi: 10.1016/j.preteyeres.2020.100918 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Maaijwee K, Heimann H, Missotten T, Mulder P, Joussen A, van Meurs J. Retinal pigment epithelium and choroid translocation in patients with exudative age-related macular degeneration: long-term results. Graefes Arch Clin Exp Ophthalmol. Nov 2007;245(11):1681–9. doi: 10.1007/s00417-007-0607-4 [DOI] [PubMed] [Google Scholar]
- 81.Heussen FM, Fawzy NF, Joeres S, et al. Autologous translocation of the choroid and RPE in age-related macular degeneration: 1-year follow-up in 30 patients and recommendations for patient selection. Eye (Lond). Jun 2008;22(6):799–807. doi: 10.1038/sj.eye.6702823 [DOI] [PubMed] [Google Scholar]
- 82.Eandi CM, Giansanti F, Virgili G. Macular translocation for neovascular age-related macular degeneration. Cochrane Database Syst Rev. Oct 8 2008;(4):Cd006928. doi: 10.1002/14651858.CD006928.pub2 [DOI] [PubMed] [Google Scholar]
- 83.Thomas BB, Lin B, Martinez-Camarillo JC, et al. Co-grafts of Human Embryonic Stem Cell Derived Retina Organoids and Retinal Pigment Epithelium for Retinal Reconstruction in Immunodeficient Retinal Degenerate Royal College of Surgeons Rats. Front Neurosci. 2021;15:752958. doi: 10.3389/fnins.2021.752958 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Anderson G, Bagnaninchi P, Dhillon B. Chapter 7 - Induced Pluripotent Stem Cells (iPSC) in Age-related Macular Degeneration (AMD). In: Birbrair A, ed. Recent Advances in iPSCs for Therapy, Volume 3. Academic Press; 2021:155–172. [Google Scholar]
- 85.Kashani AH, Lebkowski JS, Rahhal FM, et al. One-Year Follow-Up in a Phase 1/2a Clinical Trial of an Allogeneic RPE Cell Bioengineered Implant for Advanced Dry Age-Related Macular Degeneration. Transl Vis Sci Technol. Aug 12 2021;10(10):13. doi: 10.1167/tvst.10.10.13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Mandai M, Kurimoto Y, Takahashi M. Autologous Induced Stem-Cell-Derived Retinal Cells for Macular Degeneration. N Engl J Med. Aug 24 2017;377(8):792–793. doi: 10.1056/NEJMc1706274 [DOI] [PubMed] [Google Scholar]
- 87.Maeda T, Mandai M, Sugita S, Kime C, Takahashi M. Strategies of pluripotent stem cell-based therapy for retinal degeneration: update and challenges. Trends Mol Med. May 2022;28(5):388–404. doi: 10.1016/j.molmed.2022.03.001 [DOI] [PubMed] [Google Scholar]
- 88.Liu Z, Parikh BH, Tan QSW, et al. Surgical Transplantation of Human RPE Stem Cell-Derived RPE Monolayers into Non-Human Primates with Immunosuppression. Stem Cell Reports. Feb 9 2021;16(2):237–251. doi: 10.1016/j.stemcr.2020.12.007 [DOI] [PMC free article] [PubMed] [Google Scholar]