What are the therapeutic candidates targeting VEGF-A?

11 March 2025
Introduction to VEGF-A
Vascular endothelial growth factor A (VEGF-A) is a pivotal signaling protein that drives angiogenesis, the development of new blood vessels from preexisting vasculature. It is expressed in many tissues and plays a central role in both physiological and pathological processes. VEGF-A stimulates endothelial cell proliferation, migration, and survival, while also increasing vascular permeability and modulating immune responses. Because of its central role in blood vessel formation—which in turn supports tumor growth, metastasis, and the progression of ocular and inflammatory diseases—VEGF-A has become one of the most highly validated targets in modern therapy. Its importance is underscored by extensive clinical and preclinical research, leading to a range of therapeutic candidates that either block its activity directly or interrupt its downstream signaling pathways.

Role of VEGF-A in Pathophysiology
VEGF-A is not only essential for normal embryonic vasculogenesis and the maintenance of vascular integrity in adulthood, but it also becomes dramatically upregulated in response to hypoxia, cytokine stimuli, and tissue injury. In the context of cancer, hypoxic tumor regions secrete greater amounts of VEGF-A, thereby promoting neovascularization that supplies nutrients and oxygen to the rapidly proliferating tumor mass. Similarly, in ocular disorders such as neovascular age-related macular degeneration (AMD) and diabetic retinopathy, pathological overexpression of VEGF-A leads to aberrant vessel growth, leakage, and ultimately vision loss. In inflammatory and ischemic conditions, VEGF-A also plays a dual role by modulating immune cell recruitment and vascular permeability, making it central to the pathogenesis of diseases ranging from arthritis to myocardial ischemia.

Importance of VEGF-A as a Therapeutic Target
Given its diverse roles in angiogenesis and vascular permeability, targeting VEGF-A represents a promising therapeutic strategy across multiple disease domains. The rationale behind targeting VEGF-A is based on its position as a master regulator—interference with VEGF-A can disrupt the vascular support required by tumors, stabilize abnormal vessels in the retina, and reduce the deleterious inflammatory responses that compromise organ function. The fact that VEGF-A inhibition can simultaneously block multiple pathways—such as endothelial proliferation, nitric oxide production, and inflammatory cytokine release—further emphasizes its potential, as does the successful clinical experience of VEGF-A–targeted agents. As research and clinical evidence have accumulated over the past decade, VEGF-A has emerged as a validated target for both cancer and ocular diseases, leading to the development of a variety of therapeutic candidates that operate through different mechanisms of action.

Current Therapeutic Candidates Targeting VEGF-A
The therapeutic landscape for VEGF-A has expanded considerably, incorporating both established and newer agents. Current therapies include several antibody‐based agents, receptor decoys, aptamers, and small‐molecule inhibitors – each designed to block VEGF-A activity by either binding to it directly, preventing receptor engagement, or interfering with intracellular signaling processes. The synapse database provides extensive documentation of these agents, lending credence to the robust amount of preclinical and clinical data available.

Approved Therapies
Among the approved therapies targeting VEGF-A, several monoclonal antibodies and fusion proteins have been most successful.

Bevacizumab (Trade name: Avastin) is arguably the most widely recognized anti–VEGF-A monoclonal antibody. It binds to all isoforms of VEGF-A, preventing their interaction with VEGFR-1 and VEGFR-2 on endothelial cells. Bevacizumab has been approved for the treatment of various cancers—including metastatic colorectal cancer, non-small cell lung cancer, renal cell carcinoma, and glioblastoma—and is also used off-label in some ocular conditions. The mechanism behind bevacizumab’s efficacy is its ability to neutralize circulating VEGF-A, thereby inhibiting angiogenesis, reducing vascular permeability, and normalizing tumor vasculature.

Ranibizumab (Trade name: Lucentis), another antibody fragment derived from bevacizumab, has been approved primarily for intraocular use in conditions such as neovascular AMD and diabetic macular edema. It is specifically engineered for ocular penetration and rapid clearance from systemic circulation, which helps minimize systemic adverse events. Its approval by regulatory bodies such as the FDA and EMA underscores its clinical efficacy in halting and reversing vascular leakage and neovascular damage.

Aflibercept (also known as ziv-aflibercept, Trade name: Eylea in the ocular formulation) is a soluble decoy receptor fusion protein that comprises portions of VEGFR-1 and VEGFR-2 fused to an IgG Fc fragment. It binds VEGF-A with high affinity and prevents its interaction with native cell surface receptors. Aflibercept is used both in oncology and ophthalmology, reflecting its versatility in inhibiting pathologic neovascularization in a variety of disease states.

Pegaptanib sodium (Trade name: Macugen) was one of the first anti-VEGF agents approved for ocular use. Unlike the aforementioned broad-spectrum inhibitors, pegaptanib selectively binds to the VEGF165 isoform, one of the most potent mediators of pathologic angiogenesis. Although its use has diminished with the advent of ranibizumab and aflibercept, it still represents an important concept in the selective targeting of specific VEGF-A isoforms.

Conbercept is another VEGF receptor fusion protein developed in China that targets VEGF-A, VEGF-B, and placental growth factor. It is structurally similar to aflibercept but with a broader binding profile, and it is approved for ocular use, particularly in China, to treat neovascular AMD and related retinopathies.

Mechanisms of Action
The approved therapeutic candidates targeting VEGF-A share the fundamental mechanism of intercepting VEGF-A before it can activate its receptors on endothelial cells. This blockade is achieved through different molecular interactions:
- Monoclonal antibodies such as bevacizumab and ranibizumab neutralize VEGF-A by binding directly to it. This steric hindrance prevents VEGF-A from engaging VEGFR-1 and VEGFR-2, thereby diminishing the downstream proangiogenic signaling cascade.
- Fusion proteins like aflibercept and conbercept act as decoy receptors. They possess the extracellular ligand-binding domains of VEGFRs, which competitively bind VEGF-A with high affinity. This prevents VEGF-A from binding to its natural receptors on the cell membrane and thereby inhibits receptor activation and subsequent intracellular signaling.
- Pegaptanib, a ribonucleic aptamer, binds selectively to the VEGF165 isoform. Its nucleotide-based structure enables it to form a specific three-dimensional conformation that fits the receptor-binding site of VEGF165, thereby blocking its biological activity.

These mechanisms converge on the prevention of receptor tyrosine kinase activation. When VEGF-A is not available to bind, there is a downstream decrease in the activation of pathways such as PI3K/Akt, Ras/MAPK, and focal adhesion kinase cascades. These pathways are crucial for endothelial cell survival, proliferation, and migration. The net result is reduced angiogenesis, vascular leakage, and even normalization of abnormal vascular structures in tumors, which further enhances the delivery of other chemotherapies.

Clinical Efficacy and Safety Profiles
The clinical efficacy of anti–VEGF-A therapies has been demonstrated in multiple large-scale clinical trials and through routine clinical practice. For instance, bevacizumab has shown improved overall survival (OS) and progression-free survival (PFS) in various cancers when used in combination with chemotherapy. In ophthalmology, ranibizumab and aflibercept have each demonstrated significant improvements in visual acuity and reduction in retinal fluid in neovascular AMD and diabetic macular edema. Despite these successes, these agents are not without side effects.

Safety profiles vary among the agents due largely to their molecular structures and routes of administration. Systemically delivered agents like bevacizumab can cause hypertension, thromboembolic events, and gastrointestinal perforations, which highlights the need for careful patient monitoring and dose adjustments. In contrast, intraocular agents such as ranibizumab and aflibercept have a lower incidence of systemic adverse events, though ocular adverse events such as endophthalmitis, increased intraocular pressure, and retinal detachment remain considerations. Pegaptanib, with its selective inhibition, theoretically offers a reduced risk of systemic toxicity but has been largely superseded by more potent agents that provide broader inhibition of VEGF-A activity. The overall therapeutic benefits must always be balanced against these potential risks, and the clinical context often guides the choice of agent.

Emerging Therapies and Research
The research landscape for VEGF-A targeting continues to evolve, fueled by ongoing advances in molecular biology, drug design, and a deeper understanding of VEGF-A’s role in various diseases. New therapeutic candidates are emerging that seek to overcome the limitations of the current agents, particularly in terms of dosing frequency, safety, and resistance mechanisms.

Novel Drug Candidates
Innovative approaches from the synapse database include second-generation agents and new modalities such as aptamers and bispecific antibodies. One promising strategy is the development of anti–VEGF-A aptamers. Aptamers are short oligonucleotides that fold into specific three-dimensional structures capable of binding to target proteins with high affinity and specificity. These molecules can inhibit VEGF-A by binding its receptor-interacting domain, showing promise especially in ocular conditions, where localized delivery may minimize systemic exposure.

Furthermore, bispecific antibodies are being explored, which can simultaneously target VEGF-A and another receptor or ligand involved in tumor growth. For example, a bispecific molecule may target both VEGF-A and the insulin-like growth factor receptor (IGF1R), thereby blocking two complementary pathways that promote tumor angiogenesis and growth. Such therapies offer the potential to overcome resistance mechanisms that often emerge when targeting VEGF-A alone.

Another class of emerging candidates are multi-targeted tyrosine kinase inhibitors (TKIs) that include VEGF receptor blockade as part of a broader inhibitory profile. While these agents are not exclusively selective for VEGF-A, their efficacy in reducing angiogenesis and tumor progression makes them critical in combination regimens. Emerging compounds in this category continue to be refined for improved specificity and reduced off-target effects.

Preclinical and Clinical Development Stages
Many of the novel candidates described are in various stages of preclinical evaluation or early phase clinical trials. In preclinical models, these candidates have shown promising results in inhibiting tumor angiogenesis and reducing vascular permeability. For instance, studies using animal models and in vitro vascular assays have demonstrated that VEGF-A aptamers, when administered locally, can effectively suppress neovascularization with limited systemic toxicity.

Early phase clinical trials are currently assessing the safety, pharmacokinetic properties, and preliminary efficacy of these new agents. Some of these trials focus on determining the dosing regimens that will maximize VEGF-A inhibition while minimizing adverse effects. In addition to standalone trials, combination studies are being designed to assess whether these novel agents can enhance the efficacy of existing therapies such as chemotherapy or immunotherapy. These combination strategies are particularly relevant for tumors that have developed resistance to first-line anti–VEGF-A therapies.

Advances in drug delivery technologies—for example, implantable devices or sustained-release formulations—are also in development to improve patient compliance and reduce the frequency of dosing. Preclinical experiments with long-acting formulations of VEGF-A inhibitors have shown sustained suppression of angiogenic activity in animal models of ocular neovascularization, which could translate into improved clinical outcomes. Moreover, next-generation sequencing and biomarker studies are being integrated into clinical trials to identify patient populations that are most likely to respond to these emerging therapies, thereby enabling personalized treatment approaches.

Challenges and Future Directions
Despite the significant progress made in developing therapies that target VEGF-A, several challenges persist. The limitations of current therapies range from adverse safety profiles and resistance mechanisms to issues of target selectivity and dosing convenience. Addressing these challenges through innovative research will be essential for the next generation of VEGF-A–targeted treatments.

Limitations of Current Therapies
A primary limitation of currently approved agents lies in their potential for systemic toxicity and adverse events. For instance, while bevacizumab effectively neutralizes VEGF-A, its systemic administration has been associated with hypertension, thromboembolic events, and impaired wound healing. These adverse events limit the patient population that can safely receive this therapy and necessitate rigorous monitoring protocols. In the case of intraocular therapies such as ranibizumab and aflibercept, although systemic exposure is minimized, repeated injections are often required, which can lead to complications such as endophthalmitis, retinal detachment, and intraocular inflammation.

Drug resistance is another major challenge. Tumors often develop adaptive responses to VEGF-A inhibition, including the upregulation of alternative proangiogenic pathways (such as fibroblast growth factor [FGF], platelet-derived growth factor [PDGF] and placental growth factor [PlGF]) and recruitment of bone marrow–derived proangiogenic cells. This compensatory mechanism can drive treatment failure over time and is a major focus of current research aiming to identify combination strategies.

The selectivity of inhibition is also a concern. Broad-spectrum inhibitors that block all VEGF-A isoforms may inadvertently suppress normal physiological angiogenesis and impair tissue repair processes. For example, while pegaptanib’s selective inhibition of the VEGF165 isoform was intended to limit adverse effects, the overall efficacy in suppressing pathologic angiogenesis was less than that achieved by more comprehensive inhibitors like ranibizumab or aflibercept.

Patient-specific variability, including genetic polymorphisms in VEGF-A and other angiogenic factors, further complicates therapy. Biomarker studies have demonstrated that the levels of circulating VEGF-A—and the relative expression of its receptors—can differ significantly among patients, affecting both efficacy and toxicity profiles. Such heterogeneity underscores the need for personalized or stratified therapeutic approaches in clinical practice.

Future Research Directions and Innovations
Future research is geared toward overcoming these limitations by innovative approaches that combine targeted therapy with advanced drug delivery systems, novel molecular designs, and patient-specific tailoring. One promising avenue is the development of combination therapies that target multiple angiogenic pathways. For instance, bispecific antibodies that concurrently target VEGF-A and another growth factor receptor, such as IGF1R, may prevent the compensatory upregulation of alternative proangiogenic signals and overcome resistance.

Research is also focusing on developing next-generation VEGF-A inhibitors with improved pharmacokinetic profiles and reduced adverse effects. Novel candidates such as anti–VEGF-A aptamers provide the dual benefit of high specificity and potentially lower immunogenicity. In preclinical studies, these aptamers have demonstrated prolonged activity and rapid clearance when administered via local routes, which could minimize systemic exposure and related toxicity. Advanced drug delivery platforms, including sustained-release ocular implants and nanoparticle-based systems, are being explored to decrease the injection frequency required for intraocular therapies. Such innovations are expected to improve patient compliance and reduce the overall risk of injection-related complications.

In addition, researchers are investigating the therapeutic potential of selectively targeting VEGF-A isoforms. By sparing isoforms that contribute to normal physiological angiogenesis while inhibiting those that drive pathology (for example, selectively targeting the VEGF165 isoform), there is potential to maintain vascular health and tissue repair while suppressing unwanted neovascularization. This approach demands more refined molecular tools and a deeper understanding of the isoform-specific functions of VEGF-A in different tissues.

Furthermore, the integration of molecular diagnostics with therapeutic development is anticipated to enhance the precision of VEGF-A–targeted treatments. Biomarker research, which looks at circulating levels of VEGF-A, receptor expression patterns, and genetic polymorphisms, is essential for identifying patients who are most likely to benefit from these therapies. Personalized treatment regimens based on biomarker-guided patient stratification could vastly improve the therapeutic index of VEGF-A inhibitors, ensuring that patients receive the most appropriate and effective treatment.

Lastly, the development of multi-targeted kinase inhibitors that include VEGF receptor inhibition as part of a broader profile offers another promising strategy. These drugs are designed to inhibit multiple signaling pathways simultaneously, reducing the likelihood of compensatory angiogenic responses and improving overall antitumor efficacy. However, achieving the right balance between efficacy and toxicity remains a challenge, and further research is required to optimize these agents.

Conclusion
In summary, therapeutic candidates targeting VEGF-A represent a diverse and evolving class of drugs that have transformed the treatment landscape in oncology and ophthalmology. The currently approved therapies—including monoclonal antibodies such as bevacizumab and ranibizumab, as well as fusion proteins like aflibercept and conbercept, and even selective inhibitors like pegaptanib—have demonstrated considerable clinical efficacy by neutralizing VEGF-A, thereby disrupting essential angiogenic signals. Their mechanisms of action largely rely on binding to VEGF-A and preventing its interaction with its natural receptors on endothelial cells, ultimately leading to reduced endothelial proliferation, vascular leakage, and normalization of aberrant vasculature.

Emerging therapies are focused on mitigating the limitations observed with current treatments, such as adverse systemic effects, resistance mechanisms, and the need for frequent dosing. These next‐generation agents include anti–VEGF-A aptamers, bispecific antibodies that target VEGF-A alongside other contextually relevant pathways, and multi‐targeted TKIs with improved specificity and pharmacokinetic profiles. Furthermore, advances in drug delivery systems and biomarker‐guided treatment approaches promise to enhance the long‐term efficacy and safety of VEGF-A–targeted therapies.

Despite these advances, challenges remain – including managing adverse events, overcoming drug resistance, ensuring selectivity for pathological over physiological angiogenesis, and accounting for patient‐to‐patient variability. Future research is dedicated to refining these therapeutic strategies through combination regimens, molecular innovations, and personalized medicine approaches. The continued evolution of VEGF-A–targeted therapy holds great promise for improving outcomes across a broad range of neovascular and proliferative diseases.

In conclusion, the therapeutic candidates targeting VEGF-A encompass both well‐established, clinically approved agents and a wide array of novel compounds currently in the development pipeline. These candidates have demonstrated significant promise in their ability to modulate pathological angiogenesis, yet their full potential will be realized only through overcoming current limitations and embracing new technologies and personalized treatment strategies. The future of VEGF-A–targeted therapy is likely to be defined by these innovative approaches, which aim to enhance efficacy, reduce toxicity, and provide more durable clinical benefits for patients with cancer, ocular diseases, and other conditions characterized by pathological neovascularization.

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