What drugs are in development for Age Related Macular Degeneration?

12 March 2025
Overview of Age Related Macular DegenerationDefinitionon and Types
Age‐Related Macular Degeneration (AMD) is a progressive neurodegenerative disease of the retina and is one of the leading causes of irreversible central vision loss in older adults. Clinically, AMD is divided into two broad groups: dry (non‐exudative) AMD—which is characterized by the presence of drusen (yellowish deposits under the retinal pigment epithelium) and pigmentary changes—and wet (neovascular or exudative) AMD, in which abnormal blood vessels grow under the macula leading to leakage, hemorrhage, and ultimately fibrosis. Dry AMD, particularly in its advanced form known as geographic atrophy (GA), affects millions of people worldwide and can be asymptomatic for long periods. In contrast, wet AMD, which represents about 10–15% of all cases, often leads to rapid visual loss if not treated promptly. Genetic factors, including variants in the complement pathway genes (e.g., CFH) and environmental factors (such as oxidative stress and inflammation), are understood to contribute to the pathogenesis of AMD. Different stages—from early to intermediate and finally advanced AMD—are characterized not only by drusen load but also by structural and functional changes in the retinal pigment epithelium (RPE) and neuroretina, which ultimately have clinical correlations with the level of visual impairment.

Current Treatment Options
At present, the mainstay of AMD therapy—particularly for the neovascular form—remains intravitreal anti‐vascular endothelial growth factor (anti-VEGF) injections. Drugs such as ranibizumab, aflibercept, and brolucizumab (and, off-label, bevacizumab) have dramatically improved visual outcomes in patients with wet AMD by targeting VEGF and reducing angiogenesis and vascular leakage. For dry AMD, however, no approved medical therapies exist that significantly modify the course of the disease. Nutritional supplementation, based on evidence from the Age‐Related Eye Disease Study (AREDS and AREDS2), is currently recommended to slow progression in certain intermediate cases. Nevertheless, these strategies play only a supportive role and do not address the underlying biological drivers of degeneration.

Drug Development Pipeline for AMD

Preclinical and Clinical Stages
Drugs in development for AMD occupy a broad range of stages—from early preclinical research to advanced phase III clinical trials. Ongoing investigations are addressing both neovascular and non‐neovascular forms of the disease, with efforts to modulate key pathways involved in angiogenesis, the complement cascade, inflammation, oxidative stress, and even cell replacement strategies.

In the anti‐VEGF space, novel candidates include improved formulations and gene therapy approaches that are designed to produce sustained intraocular levels of anti‐VEGF proteins from a single injection. For example, RGX-314 is one gene therapy candidate that uses an adeno-associated viral (AAV) vector to drive the sustained intraocular expression of an anti‐VEGF protein, thereby potentially reducing treatment burden. In parallel, new biologic agents such as faricimab—a bispecific antibody that targets both VEGF-A and angiopoietin-2—have advanced through phase III trials and are now altering treatment paradigms by potentially extending dosing intervals in wet AMD.

Complement inhibitors represent one of the most exciting areas in the pipeline for dry AMD. Pegcetacoplan (APL-2) is a C3 inhibitor that has shown promise in reducing geographic atrophy lesion growth in phase III trials (DERBY and OAKS). Other complement pathway–modulating agents include avacincaptad pegol (also known as Zimura), which targets complement factor C5, and several factor D inhibitors that are in earlier stages of testing. Research is also being conducted on small molecules and antibodies designed to modulate other complement components and pathways—for example, inhibitors that block the activity of complement factor B or regulators that attempt to maintain a balanced complement response.

Novel approaches beyond anti-VEGF and complement inhibition include agents targeting oxidative stress and mitochondrial dysfunction, which are important in the pathogenesis of AMD. Mitochondria-targeting peptides (such as MTP-131, sometimes called Bendavia) and antioxidant compounds are under development to protect RPE cells from the oxidative and metabolic stresses that lead to degeneration. In addition, RNA-based therapies, including modified mRNA formulations and antisense oligonucleotides, are being explored to modulate gene expression and correct aberrant splicing, potentially targeting both protective and pathogenic mechanisms in AMD. Stem cell therapies also form an important part of the research landscape, particularly for the advanced stages of dry AMD. Trials investigating the transplantation of RPE cells derived from human embryonic stem cells (hESCs) or induced pluripotent stem cells (iPSCs) aim at regenerating damaged tissue and preserving or even restoring visual function.

Key Players and Companies
Several pharmaceutical companies, biotech startups, and academic groups are actively engaged in the development of AMD therapies. In the anti-VEGF arena, major companies include Genentech (ranibizumab) and Regeneron (aflibercept), but newer entrants include Novartis, which is already developing products such as brolucizumab and gene therapy candidates like RGX-314. In the field of complement inhibition, Apellis Pharmaceuticals is a notable player behind pegcetacoplan, while Iveric Bio is developing avacincaptad pegol—both are among the leaders in targeting dry AMD. Roche and its associated entities (including Genentech) continue to make significant contributions in both the investigation of anti-VEGF agents and complementary pathways. Other key players include 4D Molecular Therapeutics, which is developing sophisticated gene therapy platforms (such as the 4D-150 candidate that combines a unique intravitreal vector with an RNA interference element targeting multiple VEGF family members). In addition, academic-industry collaborations and regulation-supported consortia (like those funded by NIH and other research agencies) have notably contributed to advancing novel strategies for both VEGF-related and alternative targets in AMD.

Mechanisms of Action

Anti-VEGF Therapies
Anti-VEGF drugs remain the gold standard for neovascular AMD. However, developments in this area are not limited to repeated intravitreal injections of conventional antibodies. Newer anti-VEGF strategies include:

• Gene therapy approaches that deliver constructs encoding for anti-VEGF proteins. An example is RGX-314, which uses viral vectors to transduce retinal cells to produce a consistent level of anti-VEGF protein, thereby reducing the frequency of injections and ensuring prolonged receptor blockage.

• Improved bispecific antibodies, such as faricimab, which target not just VEGF-A but also other molecules like angiopoietin-2. This dual-action strategy is designed to not only inhibit neovascularization but also stabilize vascular permeability and reduce inflammation.

• There are also developments in sustained-release formulations, where conventional anti-VEGF antibodies are encapsulated or delivered via novel devices that allow for a slower, more constant release directly in the eye. Each of these technologies seeks to reduce the treatment burden and improve patient outcomes.

Complement Inhibitors
Complement inhibition is emerging as a promising mechanism for dry AMD and geographic atrophy. The complement system is a part of the innate immune response, and its dysregulation has been strongly associated with AMD pathogenesis. Notable approaches include:

• Pegcetacoplan (APL-2) specifically inhibits complement component C3, preventing its cleavage into pro-inflammatory fragments. Clinical trials (DERBY and OAKS) have documented reductions in the progression rate of GA lesions, reinforcing the role of complement activation in dry AMD.

• Avacincaptad pegol (Zimura) targets C5, another key protein in the cascade. By blocking C5 cleavage, it aims to avoid the formation of the membrane attack complex (MAC) and reduce downstream inflammation and cellular damage—an approach that is particularly vital in early intervention for dry AMD.

• Other investigational agents target factor D—an enzyme that initiates the alternative pathway of complement activation. Small molecules or antibodies that inhibit factor D are being explored in early-phase studies, with the goal of achieving a balanced inhibition that preserves necessary host-defense activity while reducing pathological complement overactivation.

Other Novel Approaches
Novel approaches in AMD drug development extend beyond the well-established VEGF and complement pathways. These include:

• Oxidative stress modulation and mitochondrial protection: Given that aging and oxidative stress contribute significantly to RPE cell dysfunction and death, several agents are in development that target mitochondrial dysfunction. Mitochondria-targeted antioxidants (like MTP-131) and other agents aimed at restoring mitochondrial homeostasis are in preclinical or early clinical stages.

• RNA-based therapies: Advances in modified mRNA and antisense oligonucleotide technologies have made RNA-based therapeutics an attractive option. Such therapies could regulate gene expression relevant to both protective (such as upregulation of antioxidant enzymes) and pathogenic processes (including complement dysregulation). Research into non-coding RNAs as biomarkers and therapeutic targets for AMD is also ongoing, providing insights into novel regulatory mechanisms that can be leveraged for treatment.

• Stem cell and regenerative therapies: For patients with advanced geographic atrophy or severe RPE loss, cell replacement strategies offer hope for restoring retinal function. Clinical trials employing hESC-derived or iPSC-derived RPE cells aim to replace damaged tissue and promote retinal regeneration. Although challenges remain—such as ensuring cell survival, integration, and avoiding immunogenicity—this area has seen significant investment and promising preliminary results.

• Combination therapies: Recognizing the multifactorial nature of AMD, researchers are increasingly exploring combination regimens that target two or more pathways simultaneously. For example, combining an anti-VEGF agent with a complement inhibitor or an antioxidant agent may yield synergistic effects that surpass monotherapy outcomes. Innovative clinical trial designs, including adaptive and basket trials, are being used to evaluate the efficacy of such combinations in a precision medicine framework.

Challenges and Future Directions

Research and Development Challenges
Despite the significant progress in understanding the biological basis of AMD and developing a range of therapeutic candidates, several challenges have emerged in the drug development process:

• Long-term safety and tolerability remain an important concern for intraocular treatments. Many anti-VEGF therapies require repeated intravitreal injections, which can lead to cumulative risks such as endophthalmitis, increased intraocular pressure, and patient burden. Gene therapy approaches, while promising in reducing treatment frequency, must overcome uncertainties surrounding long-term expression and immunologic responses.

• Complement inhibitors must strike a delicate balance between reducing pathogenic inflammation and preserving essential immune functions. Over-inhibition may result in heightened susceptibility to infections or unintended systemic effects. Determining the optimal dose and treatment window for these agents is an ongoing challenge.

• For novel modalities such as RNA-based therapeutics and stem cell treatments, delivery remains a key hurdle. The instability of RNA in physiological environments and the need for efficient, targeted delivery systems are significant obstacles to clinical translation, while stem cell therapies have raised concerns regarding cell differentiation, integration, and the potential for adverse events.

• The heterogeneity of AMD, both genetically and phenotypically, necessitates the identification of robust biomarkers that can predict who will benefit from specific therapies. Stratification of patients in clinical trials based on risk factors (e.g., complement gene variants) or disease stage is critical to improving trial outcomes and ultimately translating these therapies into clinical practice.

• The time and expense involved in the clinical development of new treatments for AMD are considerable. Drug development costs for novel biologics, gene therapies, and regenerative products can be in the billions of dollars, and the regulatory pathway is often long and complex. These factors place significant pressure on companies and research institutions to demonstrate both efficacy and a lowered treatment burden.

Future Prospects and Innovations
Despite these challenges, the future of AMD therapeutics appears promising with several innovative directions:

• Combination and multi-target strategies are likely to play an increasingly vital role. Rather than depending on a single target approach, future regimens may combine anti-VEGF, complement inhibition, and antioxidant therapies to provide holistic treatment that addresses the multifactorial nature of AMD. The use of adaptive clinical trial designs that allow assessment of different combination regimens could streamline this process.

• Gene therapy approaches offer the possibility of a “one‐hit” treatment that could continuously supply therapeutic proteins (such as anti-VEGF or complement inhibitors) over long periods. With newer vector designs and improved safety profiles, these therapies might dramatically reduce treatment frequency while maintaining stable intraocular drug levels.

• The advent of RNA-based drugs opens up opportunities for precision medicine in AMD. Modified mRNA, antisense oligonucleotides, and RNA interference strategies can be tailored to control the expression of multiple genes simultaneously. As these platforms mature—with enhanced stability and targeted delivery—they could offer customized treatment options for patients based on their genetic and environmental risk profile.

• Stem cell and regenerative medicine hold promise particularly for advanced dry AMD where tissue loss is significant. Ongoing clinical trials utilizing hESC-derived or iPSC-derived RPE cells have already demonstrated initial safety and feasibility, and future innovations may improve cell survival, integration, and long-term function. Parallel efforts in understanding the inflammatory milieu and ensuring appropriate immunosuppression or using autologous/allogeneic cell sources will be paramount for clinical success.

• Finally, advances in artificial intelligence (AI) and imaging have the potential to support drug development by improving patient stratification, monitoring treatment responses, and identifying new biomarkers. Standardization of imaging protocols and integration of high-resolution imaging data with clinical outcomes can help refine endpoints in clinical trials. These tools are essential for predicting which patients are most likely to progress and assessing the efficacy of new therapies early in the treatment course.

In summary, the AMD drug development pipeline is diversifying considerably. On one end, refinement of anti-VEGF therapies through gene therapy and improved formulations is designed to ease the treatment burden in neovascular AMD. On another, the emergence of complement inhibitors like pegcetacoplan and avacincaptad pegol spearheads the hope for effective treatment of dry AMD, while novel strategies—including RNA-based therapeutics, mitochondria-targeting agents, and regenerative medicine approaches—are broadening our ability to address both the vascular and degenerative elements of the disease. Each emerging modality is being evaluated meticulously through preclinical studies and increasingly sophisticated clinical trials, with major contributions from industry giants such as Novartis, Apellis, Iveric Bio, and Roche as well as innovative biotech startups like 4D Molecular Therapeutics.

The challenges remain significant, ranging from the complexity of disease pathogenesis and the need for long-term safety data, to the logistical and regulatory hurdles inherent in introducing cutting-edge biotechnologies. However, the trends suggest that the future of AMD treatment may lie in a multi-pronged, highly personalized approach that leverages combinations of these novel mechanisms. In doing so, the aim is to not only halt disease progression but also preserve and eventually restore visual function in an aging population burdened with this debilitating condition.

Conclusion:
To conclude, research for new drugs in development for Age‐Related Macular Degeneration is multifaceted and ongoing, with promising candidates emerging across various therapeutic classes. On the anti‐VEGF front, improvements through gene therapy and novel formulations offer prolonged effects and reduced injection frequency. Complement inhibitors such as pegcetacoplan and avacincaptad pegol target the inflammatory pathways implicated in dry AMD and geographic atrophy and are entering advanced clinical stages. Meanwhile, novel modalities, including RNA-based therapeutics, antioxidant and mitochondrial protective drugs, and stem cell therapies, are being explored to address the degenerative aspects of AMD that are not amenable to current treatments. Each therapeutic candidate is supported by rigorous preclinical testing and innovative phase I–III clinical trials, with close attention given to personalized medicine approaches that factor in patient genetics, disease stage, and the complex interplay of molecular pathways underlying AMD. Cutting-edge developments in imaging and AI further enhance patient selection and treatment monitoring, potentially driving down clinical trial durations and costs. Despite formidable challenges—such as ensuring long-term safety, balancing immune function during complement inhibition, and the high-cost nature of advanced biological therapies—the integration of multiple treatment modalities into combination regimens is anticipated to lead to revolutionary improvements in AMD management. As these drugs progress through the pipeline, they are expected to contribute significantly to an era of precision ophthalmology that not only preserves vision but also improves quality of life for millions of patients worldwide.

This comprehensive approach, starting from understanding the disease to scrutinizing each candidate drug’s mechanism and the practical challenges in clinical translation, underscores the robust efforts made by the AMD research community. It indicates that the horizon of therapeutic intervention for AMD will soon be markedly expanded, ultimately bringing innovative, durable, and patient-tailored treatment options to clinical practice.

Each aspect of the development pipeline—from the anti-VEGF improvements and complement inhibitors to the novel RNA and stem cell strategies—reflects an evolution in our understanding of AMD pathogenesis. Such progress is matched by enhanced clinical trial designs and strategic partnerships among key players in the industry. Collectively, these advancements herald a promising future where integrated multi-target therapy may finally transform the landscape for both neovascular and dry AMD, ensuring that therapeutic options are not only more effective but also better aligned with the complex biology of the disease.

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成