Skip to content
Publicly Available Published by De Gruyter December 7, 2022

Stem cells for treating retinal degeneration

  • Volker Busskamp ORCID logo EMAIL logo

Abstract

The mammalian retina lacks regenerative potency to replace damaged or degenerated cells. Therefore, traumatic or genetic insults that lead to the degeneration of retinal neurons or retinal pigment epithelium (RPE) cells alter visual perception and ultimately can lead to blindness. The advent of human stem cells and their exploitation for vision restoration approaches has boosted the field. Traditionally, animal models – mostly rodents – have been generated and used to mimic certain monogenetic hereditary diseases. Of note, some models were extremely useful to develop specific gene therapies, for example for Retinitis Pigmentosa, Leber congenital amaurosis and achromatopsia. However, complex multifactorial diseases are not well recapitulated in rodent models such as age-related macular degeneration (AMD) as rodents lack a macula. Here, human stem cells are extremely valuable to advance the development of therapies. Particularly, cell replacement therapy is of enormous importance to treat retinal degenerative diseases. Moreover, different retinal degenerative disorders require the transplantation of unique cell types. The most advanced one is to substitute the RPE cells, which stabilize the light-sensitive photoreceptors. Some diseases require also the transplantation of photoreceptors. Depending on the disease pattern, both approaches can also be combined. Within this article, I briefly feature the underlying principle of cell replacement therapies, demonstrate some successes and discuss certain shortcomings of these approaches for clinical application.

RPE transplantation

The retinal pigment epithelium (RPE) consists of a single cell layer that separates the retina from the choroid. RPE cells have an essential function in maintaining vision [1]. They surround the outer segments of the photoreceptors to control retinal photo-stimulation and to isomerize and thereby recharge the chromophore retinal. In addition, the RPE ingests parts of the outer segments via phagocytosis, which contributes to the daily photoreceptor maintenance. Transport across the epithelium allows the exchange of nutrients, ions, and growth factors between the retina and choroidal capillaries. In addition, pigmented RPE can absorb scattered light and thus reduce photooxidative stress. An intact RPE also contributes to the immune-privileged function of the eye by demarcating it from the blood circulation [1]. In summary, RPE cells are essential for photoreceptor function and their dysfunction or degeneration directly affects photoreceptors and thereby vision. The idea of RPE cell replacement therapy was developed almost 40 years ago and just over 30 years ago, fetal RPE cells were already transplanted into an AMD patient for the first time [2]. Since then, an abundance of research has been constantly contributing to the improvement of this method, leading, among others, to the use of non-fetal RPE cells. In particular, the use of human stem cells brought a major breakthrough and provided a variety of protocols for RPE cell differentiation. In order to avoid allogeneic rejection, immunological interplay between the host and the transplanted cells has to be considered in the development of RPE cell replacement therapy [3]. Various clinical trials with RPE cells differentiated from human stem cells could be used for the treatment of AMD and Stargardt disease [4, 5]. Partial improvement of vision was observed in these approaches. There are already first clinical studies on stem cell-derived RPE cell replacement therapy. Essentially, two methods are distinguished: RPE cells are either injected as cell suspension or transplanted into the retina as RPE choroid sheets [6]. Both techniques require treatment with immunosuppressants for a prolonged period of time, since the cell material is foreign to the body. The first clinical trials showed great success in 18 patients [7]. Here, RPE cells were derived from human embryonic stem cells and transplanted. In addition to the improvement or constant visual acuity, the risks of undesired cell growth and rejection reactions could be excluded. Thus, the mid- to long-term safety of this method could be confirmed. In another clinical study with 12 patients, the number of cells to be transplanted was investigated in more detail, as well as the integration into the patient’s retina [8]. In this study, there was focus on cell numbers, using up to 200,000 RPE cells for transplantation. Structural and especially functional integration of the cells is particularly important to provide patients with a safe and successful treatment. A three-year follow-up study with three patients could also show excellent results in Stargardt patients after transplantation with RPE cells derived from human embryonic stem cells [9]. The long-term tolerability, safety and success of the method could be confirmed once again.

Transplantation of RPE single-cell layers could also be successfully validated in clinical trials in two AMD patients. In particular, the successful approach of the transplantation of a RPE single-cell layer on an artificial membrane into the retina give hope for a broad clinical application. The main challenge with this method is the precise placement of the membrane into the retina [4]. Several clinical trials are currently underway, the results of which may lead to new therapeutic approaches. In addition, further research is being conducted focusing on integration and functionality, and thus preservation and improvement of vision in patients. For ethical reasons, the use of human induced pluripotent stem cells is more favorable. This has already been successfully tested in one patient [5]. Of note, using hESC or hiPSC-derived RPE cells, it is important that no hyperproliferating cells, which may lead to tumors, are transplanted into the retina of patients. Therefore, long-term clinical studies with a focus on safety are essential for further clinical study designs.

Photoreceptor cell replacement

Transplantation of photoreceptors into the retina to restore vision is similarly promising [10, 11]. However, photoreceptor transplantation is particularly challenging because the synaptic connection of photoreceptors to downstream bipolar cells must occur in order to relay light information. It has been previously shown that for successful transplantation, the developmental stage of photoreceptors is enormously important for their survival and synaptic integration [12]. It has also been shown that primary photoreceptor progenitor cells, which continue to mature after transplantation into the subretinal space, are particularly suitable and can improve visual function [11]. The critical steps of preparation and purification, surgical intervention, and immunomodulation after transplantation [13] are further investigated and improved in animal models. In addition, combined methods are being developed to transplant photoreceptors together with RPE cells [14]. Several years ago, some groups independently discovered that material transfer occurs when mouse photoreceptors are transplanted into mice that still have intrinsic but dysfunctional photoreceptors left [15], [16], [17]. This transfer, presumably via nanotube-like processes [18], supports and reactivates the remaining photoreceptors in the recipient. As long as nonfunctional photoreceptors are present in the recipient, the donor cells can support them by material exchange, which is also an exciting method for restoring vision.

The required cell numbers and quality for widespread clinical application of photoreceptor cell replacement therapy are the major limitations. In addition, the use of primary photoreceptor cells derived from fetal tissues is ethically, legally, and technically difficult. Therefore, alternative methods for obtaining photoreceptors have been developed, for example, from fibroblasts or pluripotent stem cells. Fibroblasts can be differentiated into photoreceptors by using transcription factors [19] or chemical agents [20]. Similarly, human stem cells have been used to grow photoreceptors in the Petri dish [21]. However, the number of photoreceptors produced by these methods has been insufficient for transplantation.

The discovery to produce whole retinas, so-called retinal organoids, from pluripotent stem cells significantly advanced the field [22]. These retinal organoids are a prolific source of human photoreceptors, which can be used to further explore the efficacy and safety of cell replacement therapy. However, this method also requires overcoming important technological and economic challenges. Cell production for medical use requires protocols that are compliant with “Good Manufacturing Practices” (GMP). This includes the complete elimination of all animal components, as well as standardized, automated production of the cells, including efficient quality control. Some progress has already been made in the field of GMP-certified photoreceptor production [23]. However, all available protocols involve extremely long cultivation phases, requiring many manual steps by trained personnel and a huge consumption of complex media. New protocols for simplified production of retinal organoids are already being developed [24, 25], but it remains to be seen whether they are feasible for efficient and economical harvesting of therapeutic cell material. Unlike foreign donor cells, autologous cells reduce the risk of immune rejection due to human leukocyte antigen (HLA) proteins [26]. De novo reprogramming of patient stem cells and subsequent correction of the mutated gene, requires personalized medicine. This is technically feasible, but it is not economical for a general therapeutic approach. Cells from HLA “super donors” that are homozygous for an HLA allele and thus compatible with a larger patient population may provide a remedy [26]. However, these special stem cell lines have yet to be verified for the production of photoreceptor-rich retinal organoids. In principle, photoreceptor cell replacement therapy looks promising in preclinical trials, but there are still some technical hurdles to overcome for broad therapeutic application.

Conclusions

Human stem cells are important tools for experimental and clinical ophthalmology. Stem cell-derived retinal cell types [27] and organoids [28] serve as a human testbed facilitating meaningful disease modeling. Protocols for generating RPE cells are very advanced and corresponding photoreceptor protocols are emerging. There are still many technical and administrative road blocks ahead of us but they will be solved by joint efforts from the field. RPE cell therapy looks extremely promising within clinical trials and photoreceptor replacement will soon leave the preclinical stage [11]. In summary, we are in an exciting era in which stem cell technology is key for the next breakthroughs in ophthalmology.


Corresponding author: Volker Busskamp, Professor for Degenerative Retinal Diseases, University Hospital Bonn, Venusberg-Campus 1 Gebäude 5, 53127 Bonn, Germany, E-mail:

Funding source: Volkswagen Foundation

Award Identifier / Grant number: A110720

Funding source: Paul Ehrlich Foundation

Award Identifier / Grant number: BU 2974/4-1

Award Identifier / Grant number: EXC-2151-390873048

  1. Research funding: VB is supported by the Volkswagen Foundation (Freigeist—A110720), the Paul Ehrlich Foundation (Frankfurt, Germany) and the Deutsche Forschungsgemeinschaft (BU 2974/4-1, EXC-2151-390873048—Cluster of Excellence—ImmunoSensation2 at the University of Bonn).

  2. Author contributions: Single author contribution.

  3. Competing interests: The author states no conflict of interest.

  4. Informed consent: Not applicable.

  5. Ethical approval: Not applicable.

References

1. Strauss, O. The retinal pigment epithelium in visual function. Physiol Rev 2005;85:845–81. https://doi.org/10.1152/physrev.00021.2004.Search in Google Scholar PubMed

2. Algvere, PV, Berglin, L, Gouras, P, Sheng, Y. Transplantation of fetal retinal pigment epithelium in age-related macular degeneration with subfoveal neovascularization. Graefes Arch Clin Exp Ophthalmol 1994;232:707–16. https://doi.org/10.1007/bf00184273.Search in Google Scholar PubMed

3. Sugita, S, Mandai, M, Kamao, H, Takahashi, M. Immunological aspects of RPE cell transplantation. Prog Retin Eye Res 2021;84:100950. https://doi.org/10.1016/j.preteyeres.2021.100950.Search in Google Scholar PubMed

4. da Cruz, L, Fynes, K, Georgiadis, O, Kerby, J, Luo, YH, Ahmado, A, et al.. Phase 1 clinical study of an embryonic stem cell-derived retinal pigment epithelium patch in age-related macular degeneration. Nat Biotechnol 2018;36:328–37. https://doi.org/10.1038/nbt.4114.Search in Google Scholar PubMed

5. Mandai, M, Watanabe, A, Kurimoto, Y, Hirami, Y, Morinaga, C, Daimon, T, et al.. Autologous induced stem-cell-derived retinal cells for macular degeneration. N Engl J Med 2017;376:1038–46. https://doi.org/10.1056/nejmoa1608368.Search in Google Scholar PubMed

6. Falkner-Radler, CI, Krebs, I, Glittenberg, C, Povazay, B, Drexler, W, Graf, A, et al.. Human retinal pigment epithelium (RPE) transplantation: outcome after autologous RPE-choroid sheet and RPE cell-suspension in a randomised clinical study. Br J Ophthalmol 2011;95:370–5. https://doi.org/10.1136/bjo.2009.176305.Search in Google Scholar PubMed

7. Schwartz, SD, Regillo, CD, Lam, BL, Eliott, D, Rosenfeld, PJ, Gregori, NZ, et al.. Human embryonic stem cell-derived retinal pigment epithelium in patients with age-related macular degeneration and Stargardt’s macular dystrophy: follow-up of two open-label phase 1/2 studies. Lancet 2015;385:509–16. https://doi.org/10.1016/s0140-6736(14)61376-3.Search in Google Scholar

8. Mehat, MS, Sundaram, V, Ripamonti, C, Robson, AG, Smith, AJ, Borooah, S, et al.. Transplantation of human embryonic stem cell-derived retinal pigment epithelial cells in macular degeneration. Ophthalmology 2018;125:1765–75. https://doi.org/10.1016/j.ophtha.2018.04.037.Search in Google Scholar PubMed PubMed Central

9. Sung, Y, Lee, MJ, Choi, J, Jung, SY, Chong, SY, Sung, JH, et al.. Long-term safety and tolerability of subretinal transplantation of embryonic stem cell-derived retinal pigment epithelium in Asian Stargardt disease patients. Br J Ophthalmol 2021;105:829–37. https://doi.org/10.1136/bjophthalmol-2020-316225.Search in Google Scholar PubMed

10. Chacko, DM, Rogers, JA, Turner, JE, Ahmad, I. Survival and differentiation of cultured retinal progenitors transplanted in the subretinal space of the rat. Biochem Biophys Res Commun 2000;268:842–6. https://doi.org/10.1006/bbrc.2000.2153.Search in Google Scholar PubMed

11. Gasparini, SJ, Llonch, S, Borsch, O, Ader, M. Transplantation of photoreceptors into the degenerative retina: current state and future perspectives. Prog Retin Eye Res 2019;69:1–37. https://doi.org/10.1016/j.preteyeres.2018.11.001.Search in Google Scholar PubMed

12. Bartsch, U, Oriyakhel, W, Kenna, PF, Linke, S, Richard, G, Petrowitz, B, et al.. Retinal cells integrate into the outer nuclear layer and differentiate into mature photoreceptors after subretinal transplantation into adult mice. Exp Eye Res 2008;86:691–700. https://doi.org/10.1016/j.exer.2008.01.018.Search in Google Scholar PubMed

13. West, EL, Pearson, RA, Barker, SE, Luhmann, UF, Maclaren, RE, Barber, AC, et al.. Long-term survival of photoreceptors transplanted into the adult murine neural retina requires immune modulation. Stem Cells 2010;28:1997–2007. https://doi.org/10.1002/stem.520.Search in Google Scholar PubMed PubMed Central

14. Thomas, BB, Lin, B, Martinez-Camarillo, JC, Zhu, D, McLelland, BT, Nistor, G, 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. https://doi.org/10.3389/fnins.2021.752958.Search in Google Scholar PubMed PubMed Central

15. Pearson, RA, Gonzalez-Cordero, A, West, EL, Ribeiro, JR, Aghaizu, N, Goh, D, et al.. Donor and host photoreceptors engage in material transfer following transplantation of post-mitotic photoreceptor precursors. Nat Commun 2016;7:13029. https://doi.org/10.1038/ncomms13029.Search in Google Scholar PubMed PubMed Central

16. Santos-Ferreira, T, Llonch, S, Borsch, O, Postel, K, Haas, J, Ader, M. Retinal transplantation of photoreceptors results in donor-host cytoplasmic exchange. Nat Commun 2016;7:13028. https://doi.org/10.1038/ncomms13028.Search in Google Scholar PubMed PubMed Central

17. Singh, MS, Balmer, J, Barnard, AR, Aslam, SA, Moralli, D, Green, CM, et al.. Transplanted photoreceptor precursors transfer proteins to host photoreceptors by a mechanism of cytoplasmic fusion. Nat Commun 2016;7:13537. https://doi.org/10.1038/ncomms13537.Search in Google Scholar PubMed PubMed Central

18. Kalargyrou, AA, Basche, M, Hare, A, West, EL, Smith, AJ, Ali, RR, et al.. Nanotube-like processes facilitate material transfer between photoreceptors. EMBO Rep 2021;22:e53732. https://doi.org/10.15252/embr.202153732.Search in Google Scholar PubMed PubMed Central

19. Seko, Y, Azuma, N, Ishii, T, Komuta, Y, Miyamoto, K, Miyagawa, Y, et al.. Derivation of human differential photoreceptor cells from adult human dermal fibroblasts by defined combinations of CRX, RAX, OTX2 and NEUROD. Genes Cells 2014;19:198–208. https://doi.org/10.1111/gtc.12127.Search in Google Scholar PubMed PubMed Central

20. Mahato, B, Kaya, KD, Fan, Y, Sumien, N, Shetty, RA, Zhang, W, et al.. Pharmacologic fibroblast reprogramming into photoreceptors restores vision. Nature 2020;581:83–8. https://doi.org/10.1038/s41586-020-2201-4.Search in Google Scholar PubMed PubMed Central

21. Lamba, DA, Gust, J, Reh, TA. Transplantation of human embryonic stem cell-derived photoreceptors restores some visual function in Crx-deficient mice. Cell Stem Cell 2009;4:73–9. https://doi.org/10.1016/j.stem.2008.10.015.Search in Google Scholar PubMed PubMed Central

22. Eiraku, M, Takata, N, Ishibashi, H, Kawada, M, Sakakura, E, Okuda, S, et al.. Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature 2011;472:51–6. https://doi.org/10.1038/nature09941.Search in Google Scholar PubMed

23. Zhu, J, Reynolds, J, Garcia, T, Cifuentes, H, Chew, S, Zeng, X, et al.. Generation of transplantable retinal photoreceptors from a current good manufacturing practice-manufactured human induced pluripotent stem cell line. Stem Cells Transl Med 2018;7:210–9. https://doi.org/10.1002/sctm.17-0205.Search in Google Scholar PubMed PubMed Central

24. Cowan, CS, Renner, M, De Gennaro, M, Gross-Scherf, B, Goldblum, D, Hou, Y, et al.. Cell types of the human retina and its organoids at single-cell resolution. Cell 2020;182:1623–40.e34. https://doi.org/10.1016/j.cell.2020.08.013.Search in Google Scholar PubMed PubMed Central

25. Volkner, M, Zschatzsch, M, Rostovskaya, M, Overall, RW, Busskamp, V, Anastassiadis, K, et al.. Retinal organoids from pluripotent stem cells efficiently recapitulate retinogenesis. Stem Cell Rep 2016;6:525–38. https://doi.org/10.1016/j.stemcr.2016.03.001.Search in Google Scholar PubMed PubMed Central

26. Nakatsuji, N, Nakajima, F, Tokunaga, K. HLA-haplotype banking and iPS cells. Nat Biotechnol 2008;26:739–40. https://doi.org/10.1038/nbt0708-739.Search in Google Scholar PubMed

27. Zuzic, M, Striebel, J, Pawlick, JS, Sharma, K, Holz, FG, Busskamp, V. Gene-independent therapeutic interventions to maintain and restore light sensitivity in degenerating photoreceptors. Prog Retin Eye Res 2022;90:101065. https://doi.org/10.1016/j.preteyeres.2022.101065.Search in Google Scholar PubMed

28. Sharma, K, Krohne, TU, Busskamp, V. The rise of retinal organoids for vision research. Int J Mol Sci 2020;21. https://doi.org/10.3390/ijms21228484.Search in Google Scholar PubMed PubMed Central

Received: 2022-10-21
Accepted: 2022-11-16
Published Online: 2022-12-07
Published in Print: 2023-07-26

© 2022 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 27.4.2024 from https://www.degruyter.com/document/doi/10.1515/jpm-2022-0510/html
Scroll to top button