Alternative Emerging Therapies to Retinal Prostheses: Comparison
Please note this is a comparison between Version 2 by Catherine Yang and Version 1 by Kevin Yang Wu.

Retinal prostheses show promise in restoring vision, but ongoing clinical trials and high costs limit their adoption. To explore alternatives for treating retinal diseases, we present an overview of cell-based and gene-based therapies. This entry aims to determine if retinal prostheses are losing favor as a therapeutic option. Cell-based therapies, particularly using pluripotent stem cells like induced pluripotent stem cells (iPSCs) and retinal progenitor cells (RPCs), hold promise for treating retinal diseases. iPSC-derived retinal pigment epithelium (RPE) cells show potential in delaying disease progression and restoring vision loss when the RPE layer is damaged. Additionally, bone marrow stem cells (BMSCs) release beneficial factors and have the ability to migrate towards lesion sites, although full differentiation into photoreceptors is not confirmed. Gene therapy also shows promise for treating retinal diseases caused by genetic mutations. Different approaches are used depending on the type of mutation (recessive or dominant). For example. viral vectors like AAV are commonly employed due to their efficient targeting and low immunogenicity. Optogenetics is a novel approach for treating inherited retinal diseases (IRDs) by introducing photosensitive proteins (opsins) to the degenerated retina. Opsins restore function and photosensitivity to remaining retinal cells. Clinical trials are underway, but more research is needed to understand opsin properties.

  • Cell-based therapies
  • Stem cells
  • Gene therapy
  • Optogenetics
  • Pluripotent stem cells
  • Viral vectors
  • Non-viral vectors
  • Retinal pigment epithelium

Cell-Based Therapies

1. Cell-Based Therapies

Cell-based therapies utilize stem cells to replace or restore dysfunctional retinal cells, often with trophic factors to support cell survival and growth[1]. Pluripotent stem cells, such as induced pluripotent stem cells (iPSCs), hold promise for retinal diseases as they can differentiate into retinal pigment epithelium (RPE) cells that support photoreceptor function. iPSC-derived RPE cell replacement shows potential in delaying disease progression and restoring vision loss in cases where the RPE layer is damaged[2].

Bone marrow stem cells (BMSCs), including mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs), are extracted from the bone marrow. While BMSCs have more limited differentiation capacity compared to pluripotent stem cells, they can generate tissue-specific cells. Notably, BMSCs possess the ability to transdifferentiate and migrate towards lesion sites [97]. While full differentiation into photoreceptors has not been confirmed, recent preclinical studies demonstrate their release of beneficial factors, including anti-angiogenic and neurotrophic factors, insulin-like growth factor-1 (IGF-1), Th2-related cytokines, and the class II major histocompatibility complex (MHC II)[3].

Damage to the RPE layer can lead to vision loss, making stem-cell-derived RPE an intriguing treatment option for retinal diseases. RPE offers trophic support to remaining photoreceptors and its well-known morphology and physiology make it a suitable candidate for stem cell therapies. The eye's immune privilege and favorable microenvironment further enhance the feasibility of stem cell therapies[4][5]. Co-transplantation of fetal RPE and mesenchymal stem cells (MSCs) in mice demonstrated improved electroretinograms (ERGs), increased rhodopsin levels, decreased caspase-3 expression, and enhanced survival of transplanted cells. Coculture transplantation outperformed single-cell transplantation, presenting opportunities for advancing retinal disease treatments[6].

Stem-cell-based therapies hold potential for treating retinal diseases associated with RPE dysfunction and loss, such as age-related macular degeneration (AMD) and Stargardt's macular dystrophy (SMD). While safety of stem-cell-derived RPE transplantation has been confirmed, further investigation is needed to assess visual outcomes. Optimal timing of RPE transplantation, occurring after RPE dysfunction but before photoreceptor injury, is crucial for preserving photoreceptor function. However, systemic immunosuppression required for stem-cell-based therapies may limit their use in patients with comorbidities, reducing the pool of eligible candidates[5].

Retinal progenitor cells (RPCs) derived from developing human fetal neural retina show promise as another type of stem cell. RPCs express photoreceptor markers and have the ability to secrete trophic factors, supporting retinal survival and potentially enabling photoreceptor replacement[7][8][9]. Preclinical studies demonstrate the regenerative potential of stem cells, as they can differentiate RPCs into photoreceptors and establish functional synapses with the host, potentially stabilizing or reversing vision loss.

An example of successful vision restoration through stem-cell-based implants is the "California Project to Cure Blindness Retinal Pigment Epithelium" (CPCB-PRE1), where a monolayer of human embryonic stem cell-derived RPE cultured on a Bruch's membrane-mimicking media improved vision and slowed progressive vision loss in a phase I/IIa clinical study[10][11][12].

Gene-Based Therapies

2. Gene-Based Therapies

Gene therapy has emerged as a promising approach for addressing retinal diseases caused by genetic mutations, such as retinitis pigmentosa (RP). With its ability to target the root cause of the disease, gene therapy holds the potential to restore vision in affected individuals. The specific approach employed depends on whether the inherited retinal disease stems from a recessive or dominant genetic mutation.

For recessive mutations, a gene complementation approach is typically utilized, while dominant mutations are preferably addressed through gene suppression, with or without gene complementation. To enable gene therapy, scientists have developed various techniques, including the use of viral and non-viral vectors, as well as CRISPR-Cas9 gene editing. These techniques allow for targeted delivery and the expression of corrective genes within the retina[13].

Viral vectors utilized in gene therapy can be categorized into three subtypes: adenovirus (Ad), adeno-associated virus (AAV), and lentivirus (LV). Among these, AAV has garnered significant attention for treating inherited retinal diseases due to its small size (25 nm), enabling efficient targeting of the retinal layers. Additionally, AAV exhibits low immunogenicity, making it a favorable candidate for long-acting treatments[14]. However, due to its limited capacity to accommodate large genomes (packaging capacity of 4.7 kb), AAV is primarily used to treat recessive diseases such as Stargardt's disease[14].

Non-viral vectors have also been explored for gene delivery due to their reduced risk of immune responses and their ability to deliver larger genes compared to viral vectors. However, unlike viral vectors, non-viral vectors have not yet demonstrated efficient delivery of genetic material into cells, primarily due to the lack of efficient mechanisms for nucleus entry[15].

The gene-editing system known as CRISPR-Cas9 has emerged as a valuable tool for gene expression and suppression. It has shown promise in silencing dominant mutations using the non-homologous end-joining pathway in inherited retinal diseases. However, the success of CRISPR-Cas9 is highly dependent on factors such as guide RNA design, the type of target cell, the choice between viral or non-viral vectors, and host-related considerations[16].

Currently, the only approved gene therapy for treating a retinal disease is Luxturna, which targets the RPE65 gene and utilizes an AAV vector to treat Leber Congenital Amaurosis type 2 (LCA2). LCA2 is a suitable candidate for gene therapy as it involves an enzyme expressed in the RPE layer, which is more accessible for AAV targeting compared to the photoreceptor layer. However, most other inherited retinal diseases involve gene transfer to the photoreceptor layer, posing a challenge for gene therapy[15].

Despite the potential of gene therapy for treating inherited retinal diseases, its practical application is complicated by the wide variety of gene mutations causing these conditions. A comprehensive understanding of pathogenesis is necessary, and further research is required to assess the long-term outcomes and safety of this therapy[17].

Optogenetics

3. Optogenetics

Optogenetics has emerged as a new approach for treating inherited retinal diseases (IRDs). Unlike gene therapy, which aims to restore mutated genes, optogenetics involves introducing photosensitive proteins (opsins) to the degenerated retina. By doing so, it restores function and provides photosensitivity to remaining retinal cells, primarily non-light-sensitive cells like bipolar and retinal ganglion cells.

Two types of opsins are used: microbial opsins (type 1) and animal opsins (type 2), each with different light sensitivity and functions. Type 1 opsins directly affect ion channels or pumps, while type 2 opsins indirectly affect ion channels through intracellular G-protein-coupled receptor signaling cascades. The choice of opsin depends on the desired effect, mimicking either an "off" or "on" response[18][19][20].

Phase I/II clinical trials are currently underway for optogenetic therapy, building upon the established proof of concept. However, further research is still required to understand the structure, molecular transport modes, dynamics, and optical properties of photosensitive proteins[17].

Verdict

4. Verdict

When considering treatment options for inherited retinal diseases (IRDs), retinal prostheses may appear less attractive compared to cell-based and gene-based therapies. However, each approach has its strengths and challenges, and having multiple options allows for leveraging strengths to overcome weaknesses. For instance, a study by Nascimento-dos-Santos et al. demonstrated the synergistic effect of combining gene therapy (AAV2-PEDF) with cell therapy (human mesenchymal stem cell) in promoting neuroprotection and axonal outgrowth in the retinal ganglion cell (RGC) layer[21].

Optogenetics presents another intriguing solution for enhancing retinal prostheses, as highlighted by Lagali et al. Optogenetic prostheses eliminate the need for invasive surgery and reduce biocompatibility concerns. They also offer potential solutions to address the limited visual restoration achieved by retinal implants, relying on high brightness displays and appropriate optics[22]. Moreover, a study by Gongxin Li et al. demonstrated that optogenetic implants incorporating ChR2-expressing cells achieved better resolution than current implants, hinting at the possibility of a new generation of retinal prostheses[23].

While retinal prostheses face challenges in terms of vision quality and surgical risks, their combination with cell, gene, or optogenetics therapy holds promise for greater neuroprotection and improved vision restoration. Consequently, interdisciplinary collaboration among ophthalmologists, geneticists, stem cell biologists, engineers, and others is crucial to advance and refine therapeutic approaches, ensuring optimal outcomes for patients. Continued research and development of these diverse treatment avenues remain essential for the effective management of inherited retinal diseases.

Table 1 provides a comparative overview of the emerging treatment modalities for retinal diseases, including retinal prostheses, cell-based therapies, gene-based therapies, and optogenetics, highlighting their core mechanisms, potential advantages, and current limitations.
Table 1.
Comparative analysis of emerging therapies for retinal diseases.
Treatment Modality Description Advantages Limitations
Retinal Prostheses This technology works by artificially stimulating the retinal nerve cells to mimic the function of lost or damaged photoreceptors. Can restore some vision in patients with advanced retinal diseases, such as retinitis pigmentosa. Safety and efficacy are still being evaluated. High cost may limit widespread adoption.
Cell-Based Therapies Therapies involve the use of stem cells (pluripotent stem cells, bone marrow stem cells, and retinal progenitor cells) to replace or restore dysfunctional cells in the retina. Potential to delay disease progression and restore vision loss, and can provide trophic support to remaining photoreceptors. Potential risk of immune rejection.
Gene-Based Therapies Therapies involve the use of viral and non-viral vectors and CRISPR-cas9 gene editing to correct genetic mutations causing retinal diseases. Targets the root cause of the disease, potentially restoring vision. Limited by the variety of gene mutations, so long-term outcomes and safety still require further investigation.
Optogenetics This technique involves introducing photosensitive proteins to the degenerated retina to restore function and provide photosensitivity to remaining retinal cells. Can restore photosensitivity to non-light-sensitive cells. Requires further research on the structure, transport modes, dynamics, and optical properties of photosensitive proteins.

References

  1. Jindal, N.; Banik, A.; Prabhakar, S.; Vaiphie, K.; Anand, A. Alteration of Neurotrophic Factors After Transplantation of Bone Marrow Derived Lin-ve Stem Cell in NMDA-Induced Mouse Model of Retinal Degeneration. J. Cell. Biochem. 2017, 118, 1699–1711.
  2. Mandai, M.; Fujii, M.; Hashiguchi, T.; Sunagawa, G.A.; Ito, S.; Sun, J.; Kaneko, J.; Sho, J.; Yamada, C.; Takahashi, M. IPSC-Derived Retina Transplants Improve Vision in Rd1 End-Stage Retinal-Degeneration Mice. Stem Cell Rep. 2017, 8, 69–83.
  3. Bassi, Ê.J.; de Almeida, D.C.; Moraes-Vieira, P.M.M.; Câmara, N.O.S. Exploring the Role of Soluble Factors Associated with Immune Regulatory Properties of Mesenchymal Stem Cells. Stem Cell Rev. Rep. 2012, 8, 329–342.
  4. Kolomeyer, A.M.; Zarbin, M.A. Trophic Factors in the Pathogenesis and Therapy for Retinal Degenerative Diseases. Surv. Ophthalmol. 2014, 59, 134–165.
  5. Sachdeva, M.M.; Eliott, D. Stem Cell-Based Therapy for Diseases of the Retinal Pigment Epithelium: From Bench to Bedside. Semin. Ophthalmol. 2016, 31, 25–29.
  6. Pan, T.; Shen, H.; Yuan, S.; Lu, G.; Zhang, Y.; Wang, H.; Zhao, Y.; Sun, X.; Liu, Q. Combined Transplantation With Human Mesenchymal Stem Cells Improves Retinal Rescue Effect of Human Fetal RPE Cells in Retinal Degeneration Mouse Model. Investig. Ophthalmol. Vis. Sci. 2020, 61, 9.
  7. Wang, N.-K.; Tosi, J.; Kasanuki, J.M.; Chou, C.L.; Kong, J.; Parmalee, N.; Wert, K.J.; Allikmets, R.; Lai, C.-C.; Chien, C.-L.; et al. Transplantation of Reprogrammed Embryonic Stem Cells Improves Visual Function in a Mouse Model for Retinitis Pigmentosa. Transplantation 2010, 89, 911–919.
  8. Klassen, H.J.; Ng, T.F.; Kurimoto, Y.; Kirov, I.; Shatos, M.; Coffey, P.; Young, M.J. Multipotent Retinal Progenitors Express Developmental Markers, Differentiate into Retinal Neurons, and Preserve Light-Mediated Behavior. Investig. Ophthalmol. Vis. Sci. 2004, 45, 4167–4173.
  9. Stern, J.H.; Tian, Y.; Funderburgh, J.; Pellegrini, G.; Zhang, K.; Goldberg, J.L.; Ali, R.R.; Young, M.; Xie, Y.; Temple, S. Regenerating Eye Tissues to Preserve and Restore Vision. Cell Stem Cell 2018, 22, 834–849.
  10. Sharma, R.; Khristov, V.; Rising, A.; Jha, B.S.; Dejene, R.; Hotaling, N.; Li, Y.; Stoddard, J.; Stankewicz, C.; Wan, Q.; et al. Clinical-Grade Stem Cell–Derived Retinal Pigment Epithelium Patch Rescues Retinal Degeneration in Rodents and Pigs. Sci. Transl. Med. 2019, 11, eaat5580.
  11. Kashani, A.H.; Lebkowski, J.S.; Rahhal, F.M.; Avery, R.L.; Salehi-Had, H.; Dang, W.; Lin, C.-M.; Mitra, D.; Zhu, D.; Thomas, B.B.; et al. A Bioengineered Retinal Pigment Epithelial Monolayer for Advanced, Dry Age-Related Macular Degeneration. Sci. Transl. Med. 2018, 10, eaao4097.
  12. Kashani, A.H.; Uang, J.; Mert, M.; Rahhal, F.; Chan, C.; Avery, R.L.; Dugel, P.; Chen, S.; Lebkowski, J.; Clegg, D.O.; et al. Surgical Method for Implantation of a Biosynthetic Retinal Pigment Epithelium Monolayer for Geographic Atrophy: Experience from a Phase 1/2a Study. Ophthalmol. Retin. 2020, 4, 264–273.
  13. Ong, T.; Pennesi, M.E.; Birch, D.G.; Lam, B.L.; Tsang, S.H. Adeno-Associated Viral Gene Therapy for Inherited Retinal Disease. Pharm. Res. 2019, 36, 34.
  14. Trapani, I.; Puppo, A.; Auricchio, A. Vector Platforms for Gene Therapy of Inherited Retinopathies. Prog. Retin. Eye Res. 2014, 43, 108–128.
  15. Trapani, I.; Auricchio, A. Has Retinal Gene Therapy Come of Age? From Bench to Bedside and Back to Bench. Hum. Mol. Genet. 2019, 28, R108–R118.
  16. Ahmad, I. CRISPR/Cas9—A Promising Therapeutic Tool to Cure Blindness: Current Scenario and Future Prospects. Int. J. Mol. Sci. 2022, 23, 11482.
  17. Liu, W.; Liu, S.; Li, P.; Yao, K. Retinitis Pigmentosa: Progress in Molecular Pathology and Biotherapeutical Strategies. Int. J. Mol. Sci. 2022, 23, 4883.
  18. Tomita, H.; Sugano, E.; Fukazawa, Y.; Isago, H.; Sugiyama, Y.; Hiroi, T.; Ishizuka, T.; Mushiake, H.; Kato, M.; Hirabayashi, M.; et al. Visual Properties of Transgenic Rats Harboring the Channelrhodopsin-2 Gene Regulated by the Thy-1.2 Promoter. PLoS ONE 2009, 4, e7679.
  19. Simon, C.-J.; Sahel, J.-A.; Duebel, J.; Herlitze, S.; Dalkara, D. Opsins for Vision Restoration. Biochem. Biophys. Res. Commun. 2020, 527, 325–330.
  20. Deisseroth, K. Optogenetics: 10 Years of Microbial Opsins in Neuroscience. Nat. Neurosci. 2015, 18, 1213–1225.
  21. Nascimento-Dos-Santos, G.; Teixeira-Pinheiro, L.C.; da Silva-Júnior, A.J.; de Carvalho, L.R.P.; Mesentier-Louro, L.A.; Hauswirth, W.W.; Mendez-Otero, R.; Santiago, M.F.; Petrs-Silva, H. Effects of a Combinatorial Treatment with Gene and Cell Therapy on Retinal Ganglion Cell Survival and Axonal Outgrowth after Optic Nerve Injury. Gene Ther. 2020, 27, 27–39.
  22. Barrett, J.M.; Berlinguer-Palmini, R.; Degenaar, P. Optogenetic Approaches to Retinal Prosthesis. Vis. Neurosci. 2014, 31, 345–354.
  23. Li, G.; Wang, F.; Yang, W.; Yang, J.; Wang, Y.; Wang, W.; Liu, L. Development of an Image Biosensor Based on an Optogenetically Engineered Cell for Visual Prostheses. Nanoscale 2019, 11, 13213–13218.
More
Video Production Service