What's the latest update on the ongoing clinical trials related to VEGF-A?

20 March 2025
Introduction to VEGF-A
Vascular endothelial growth factor‐A (VEGF-A) is recognized as a master regulator of angiogenesis, playing a pivotal role in both physiological processes such as wound healing and embryogenesis as well as in pathologic conditions including solid tumor growth, retinal diseases, and ischemic cardiovascular conditions. VEGF-A drives the formation and remodeling of blood vessels through the stimulation of endothelial cell proliferation, migration, and survival. It also modulates vascular permeability and thereby participates in tissue repair, neovascularization, and—in the context of cancer—tumor progression via enhancement of the vascular supply to neoplastic tissues.

Role and Importance in Angiogenesis
VEGF-A is fundamental in regulating angiogenic processes. Its binding to high-affinity receptors (primarily VEGFR-2 and, to a lesser extent, VEGFR-1) initiates downstream signaling cascades (such as the MAPK and PI3K/Akt pathways) that are essential for the proliferation and survival of endothelial cells. This growth factor has become the focus of intense study because slight modifications in its expression or signaling can have profound effects on the development of new blood vessels. In pathological states such as cancer, chronic ischemia, and diabetic complications, dysregulated VEGF-A leads to aberrant vessel growth and permeability changes. Such alterations not only support disease progression but also create a microenvironment that is resistant to conventional therapies. The capacity to modulate VEGF-A signaling thus represents a critical strategy in the treatment of angiogenesis-dependent diseases.

Therapeutic Target in Diseases
Given its central role in angiogenesis, VEGF-A is an attractive therapeutic target across several disease areas. In oncology, anti-VEGF therapies have been developed to inhibit tumor angiogenesis, thereby starving the tumor of its blood supply and limiting its growth and metastatic potential. Similarly, in ocular diseases such as neovascular (wet) age-related macular degeneration (nAMD) and diabetic macular edema (DME), overexpression of VEGF-A leads to abnormal blood vessel growth and leakage, which can compromise vision. Accordingly, agents that inhibit VEGF-A or block its receptors have been effectively incorporated into standard treatment protocols for these indications. Furthermore, emerging approaches in cardiovascular medicine are exploring the potential of localized VEGF-A delivery to improve tissue perfusion and promote reparative angiogenesis in ischemic heart disease and peripheral arterial disease. These diverse strategies underscore VEGF-A’s status as a versatile target, with therapeutic interventions tailored according to the disease context and the mechanism of pathological vascular remodeling.

Current Clinical Trials Involving VEGF-A
Ongoing clinical trials are actively investigating VEGF-A both as a target for inhibition in conditions with pathological angiogenesis (such as ocular diseases and certain cancers) and as a pro-angiogenic therapeutic in situations where enhanced blood vessel formation is desired (for example, in ischemic cardiovascular diseases). These trials are employing innovative drug delivery systems and novel molecular platforms, ranging from mRNA‐based therapies to recombinant proteins and antibody traps, to modulate VEGF-A signaling with improved precision and safety.

Overview of Ongoing Trials
Several clinical trials focused on VEGF-A are currently in process, reflecting the broad therapeutic applicability of targeting this molecule. In the realm of ophthalmology, multiple Phase III and IV studies are investigating anti-VEGF-A agents. For example, the AVONELLE-X trial is evaluating the long-term safety and tolerability of Vabysmo—a therapy for neovascular age-related macular degeneration—while similar studies such as Rhone-X target diabetic macular edema. Another notable trial, ELEVATUM, is assessing the treatment response of faricimab, a dual Angiopoietin-2 and VEGF-A inhibitor, in treatment-naïve, underrepresented patients with DME. These studies typically use intravitreal injections to ensure direct delivery of the therapeutic agent into the eye, thereby maximizing local efficacy while minimizing systemic exposure.

In cardiovascular research, the development of localized VEGF-A therapies is gaining momentum. Moderna’s localized VEGF-A program, AZD8601, is a promising mRNA‐based therapeutic developed in collaboration with AstraZeneca. This agent is designed for direct administration (intramyocardial injection) in patients at risk of or suffering from ischemic heart disease and heart failure. Phase 1a/b and Phase II studies have demonstrated that AZD8601 is safe and well tolerated, shows evidence of protein production, and produces favorable changes in biomarkers such as blood flow and left ventricular ejection fraction. Although AstraZeneca has returned the rights to this program, the promising early-phase data suggest that further trials will be initiated under Moderna’s stewardship or through collaborative efforts.

Across oncology, efforts are being made to optimize the delivery of anti-VEGF-A therapies to overcome resistance mechanisms and to improve patient outcomes. Trials are investigating dosing regimens, administration routes (direct vs. systemic), and combination strategies where anti-VEGF-A agents are used alongside standard chemotherapy or immune checkpoint inhibitors. Adaptive trial designs and the incorporation of biomarker analyses are becoming standard to identify patient subgroups likely to benefit from such therapies.

Key Objectives and Designs
The key objectives of these ongoing clinical trials vary according to the therapeutic indication. In ocular trials, primary endpoints often include the safety profile as determined by adverse event frequency, anatomical efficacy measured by reduced retinal thickness, and functional outcomes such as improvement in best-corrected visual acuity. Secondary endpoints might include changes in the frequency of injections required, durability of the treatment effect, and biomarker studies evaluating VEGF-A levels in ocular fluids.

Cardiovascular trials, such as those evaluating AZD8601, primarily focus on safety and tolerability, while also assessing biological efficacy by measuring improvements in myocardial blood flow, reduction in ischemic biomarkers (e.g., NT-proBNP), and improvements in cardiac function (e.g., left ventricular ejection fraction). These trials employ innovative strategies intended to localize VEGF-A delivery directly to affected myocardial tissue, thereby harnessing its pro-angiogenic potential to stimulate reparative neovascularization.

In cancer trials, the designs are often randomized and controlled, with an emphasis on comparing standard of care treatment regimens with regimens that incorporate anti-VEGF-A agents. The endpoints focus on progression-free survival (PFS), overall survival (OS), and objective response rates (ORRs). In addition, patient selection is increasingly guided by biomarker analyses, including baseline plasma VEGF-A levels and specific VEGF-A receptor polymorphisms, which may serve as predictive indicators for response to therapy. The trials are designed to address not only the effectiveness of treatment in shrinking tumors and prolonging survival but also in minimizing adverse effects by adjusting the dosage and administration route—a factor that has been shown to influence outcomes significantly.

Latest Findings and Results
Recent updates emerging from these clinical trials indicate promising results in terms of safety, tolerability, and early signs of efficacy, although variations still exist among different treatment modalities and patient populations.

Recent Updates from Major Trials
In ocular indications, the latest data demonstrate that anti-VEGF-A therapies continue to show favorable safety profiles and promising efficacy outcomes. For instance, the AVONELLE-X trial (evaluating Vabysmo for nAMD) and the Rhone-X trial (targeting diabetic macular edema) have both shown that long-term administration via intravitreal injections does not compromise retinal function and offers sustained visual improvement with a low incidence of serious adverse effects. These trials are also emphasizing patient-reported outcomes, which have begun to indicate improved quality of life measures in addition to objective visual improvements.

Furthermore, the ELEVATUM trial is adding to the body of evidence by focusing on an underrepresented patient population with DME, aiming to verify whether faricimab (which targets both Angiopoietin-2 and VEGF-A) can provide enhanced efficacy over traditional anti-VEGF-A monotherapies. Early findings from ELEVATUM suggest a trend toward reduced treatment burden (i.e., fewer injections) and robust anatomical improvements, although final results are still pending.

In cardiovascular applications, the AZD8601 program has recently provided clinical proof-of-mechanism data. The Phase 1a/b studies revealed that local administration of AZD8601 in diabetic patients with cardiac dysfunction induces measurable increases in VEGF-A protein levels at the site of injection, resulting in improved regional blood flow. The subsequent Phase II study met its primary endpoint of safety and tolerability while also showing numerical trends toward improved cardiac function and reduced biomarker levels such as NT-proBNP. These results, although preliminary, underscore the potential for mRNA-based VEGF-A therapies to contribute to myocardial repair and highlight the importance of direct, localized administration in enhancing therapeutic outcomes.

In oncology, a growing number of studies are leveraging biomarker-driven patient selection to determine which individuals are most likely to benefit from VEGF-A inhibition. Recent meta-analyses and subgroup analyses have indicated that patients with higher baseline plasma VEGF-A may derive greater progression-free survival benefit from treatments incorporating agents like bevacizumab. Moreover, ongoing trials have incorporated stratification based on genetic polymorphisms in VEGF-A and its receptors, which may further refine therapeutic efficacy and safety profiles. Adaptive designs in these studies are now allowing for dynamic enrollment adjustments, making it possible to better target therapies to responsive subgroups without compromising overall trial integrity.

Comparative Analysis of Trial Outcomes
A comparative look at the outcomes across various clinical trials reveals that route and timing of administration are critical factors influencing the success of VEGF-A–targeted interventions. In ocular conditions, direct intravitreal injections have consistently outperformed indirect systemic administration methods. The localized delivery ensures that therapeutic levels of the anti-VEGF-A agents are maintained in the retina, leading to sustained reductions in retinal thickness and better visual acuity outcomes. In contrast, studies using systemic approaches in cardiovascular settings have shown that the proximity of drug delivery to the affected tissue (for instance, intramyocardial versus intravenous administration) significantly alters the magnitude of therapeutic benefit, with direct administration leading to greater improvements in perfusion and myocardial function.

Furthermore, comparative assessments between monotherapy and combination approaches highlight that dual-target inhibitors—such as faricimab, which simultaneously antagonizes VEGF-A and Angiopoietin-2—may provide superior outcomes in certain patient populations, particularly those with refractive or resistant forms of DME. This dual inhibition strategy addresses not only the aberrant angiogenic process driven by VEGF-A but also the complementary pro-permeability and destabilizing effects of Angiopoietin-2 on the vasculature. Early evidence from the ELEVATUM trial suggests that such combination therapies might reduce the injection frequency required, thereby lessening patient discomfort and treatment burden while maintaining efficacy.

In oncology, the heterogeneity of tumor response to VEGF-A inhibition has been a recurring theme. Trials that incorporate robust biomarker analyses have begun to unravel the complex interplay between VEGF-A levels, receptor expression, and tumor microenvironment, thereby enabling more personalized interventions. While some studies have reported significant improvements in progression-free survival in high VEGF-A expressers, others have suggested that a subset of patients may exhibit intrinsic resistance due to alternative angiogenic pathways or receptor polymorphisms. Comparative analyses thus emphasize the need for personalized treatment regimens that integrate both dose optimization and patient stratification, an approach that ongoing trials are starting to implement with promising preliminary results.

Implications and Future Directions
The latest clinical trial updates related to VEGF-A have important implications for clinical practice and future research strategies. These studies not only validate the therapeutic targeting of VEGF-A but also illuminate pathways to enhance efficacy by refining delivery methods and by combining therapies to overcome resistance mechanisms.

Clinical Implications of Trial Results
The recent data from ocular, cardiovascular, and oncology trials collectively confirm that modulation of VEGF-A can lead to significant clinical benefits. For ocular diseases such as nAMD and DME, the long-term safety and efficacy demonstrated by agents like Vabysmo and faricimab reinforce the current standard of care while also paving the way for treatments with extended durability and reduced treatment burden. The fact that these agents have maintained a favorable safety profile over prolonged studies is particularly reassuring given the chronic nature of these conditions.

In cardiovascular medicine, the promising outcomes with AZD8601 signal a possible paradigm shift from traditional protein-based therapies to mRNA-based gene therapy approaches. The ability to locally induce VEGF-A production in ischemic myocardial regions without systemic adverse effects offers considerable promise for enhancing myocardial repair in heart failure patients. As these therapies move toward later-stage clinical trials, they could potentially reduce the need for more invasive interventions by promoting endogenous tissue repair mechanisms.

Oncology trials underscore the importance of patient stratification through biomarker analysis. The identification of high baseline VEGF-A levels as a potential predictor of response to therapies such as bevacizumab supports ongoing efforts to tailor treatment strategies to individual patient profiles. These findings will likely refine clinical decision-making by enabling clinicians to select patients who are most likely to benefit from VEGF-A–targeted therapies, thus optimizing overall treatment efficacy while reducing unnecessary exposure to potential side effects.

Moreover, the integration of novel trial designs—such as adaptive and biomarker-driven studies—represents an important evolution in clinical research methodologies. These designs not only increase the efficiency of trials but also generate high-quality data that can be directly translated into clinical practice. The successful implementation of such strategies in VEGF-A trials may serve as a blueprint for future studies on other molecular targets in diverse disease settings.

Potential Future Research Directions
Future research in the field of VEGF-A therapeutics should address several key areas to further improve clinical outcomes and broaden therapeutic applications. First, optimizing the route, timing, and dosage of VEGF-A–targeted agents remains a central focus. For example, refining intravitreal injection protocols in ocular trials or improving local delivery techniques in cardiovascular studies will be essential to maximize therapeutic benefits while minimizing adverse effects. Preclinical studies have already indicated that direct administration near the target tissue can dramatically improve outcomes; translating these findings into larger, confirmatory clinical trials will be a priority.

Second, there is a need for more robust biomarker development and validation. Identifying reliable biomarkers—such as plasma VEGF-A levels, receptor polymorphisms, or even gene expression profiles within the tumor microenvironment—will not only improve patient selection but also enable dynamic monitoring of treatment response. As some trials have already indicated, patients with high baseline VEGF-A may respond differently to therapy compared with those with low expression levels. Incorporating such biomarker-driven stratification into future trial designs could help tailor individualized treatment regimens, ultimately enhancing efficacy and safety.

Third, combination therapeutic strategies hold great promise. The dual inhibition of VEGF-A and other synergistic factors (for instance, Angiopoietin-2) has already shown encouraging trends in reducing injection frequency and enhancing clinical outcomes in ocular disease. Similar combination approaches could be explored in other therapeutic areas; for example, in oncology, combining anti-VEGF-A agents with immunotherapies or chemotherapy regimens may overcome the compensatory mechanisms that often lead to therapeutic resistance. Future studies should also investigate the sequencing of therapies—determining whether concurrent or sequential administration offers superior outcomes—and explore new molecular combinations that synergize with VEGF-A inhibition.

Finally, advances in drug delivery technology, such as nanoparticle carriers, hydrogels, and sustained-release formulations, are likely to play an essential role in the next generation of VEGF-A–targeted therapies. Such technologies could facilitate more controlled release of the therapeutic agent, ensure a higher degree of localization, and ultimately lead to more consistent and durable effects. This is especially pertinent in diseases where continuous modulation of angiogenesis is required over a prolonged period, such as in chronic ocular and cardiovascular conditions.

Adaptive and innovative clinical trial designs that incorporate real-time monitoring, flexibility in dosing adjustments, and interim analyses will also be pivotal. These designs can help in rapidly identifying the most promising therapeutic regimens and discontinuing less effective ones, thereby accelerating the path from bench to bedside. Moreover, integrating artificial intelligence and data analytics into clinical trial management may further refine patient stratification and predict long-term outcomes more accurately, setting the stage for truly personalized medicine in the field of angiogenesis.

Conclusion
In summary, the latest updates on ongoing clinical trials related to VEGF-A highlight a dynamic and evolving field with significant therapeutic potential across multiple disease areas. Recent clinical trials in ophthalmology have provided robust evidence for the effectiveness and safety of direct intravitreal anti-VEGF-A therapies, demonstrating improvements in visual outcomes and reductions in treatment burden. Similarly, cardiovascular trials employing localized mRNA-based VEGF-A gene therapy like AZD8601 show promise in enhancing myocardial repair and improving cardiac function. In oncology, patient stratification based on VEGF-A biomarkers is emerging as a key strategy to maximize treatment efficacy and tailor therapy to individual patient profiles.

Moreover, the landscape of VEGF-A–targeted therapies is being reshaped by advances in combination strategies, innovative drug delivery systems, and adaptive trial designs. These efforts are aimed at optimizing the therapeutic window—balancing efficacy with safety—and at understanding the mechanisms of resistance that sometimes limit the long-term benefits of VEGF-A modulation. Future research directions include further refinement of dosing and delivery methods, the development and implementation of robust biomarkers for patient selection, and expansive studies that explore combination therapies and innovative formulations. Such comprehensive strategies, built upon the promising early data from ongoing clinical trials, are expected to not only refine the therapeutic applications of VEGF-A targeting but also set new standards in personalized medicine and adaptive clinical trial design.

Overall, the latest clinical trial updates affirm that VEGF-A remains a vital therapeutic target, with emerging therapies showing both promise and the potential for transformative clinical impact. Continued collaborative efforts between academia, industry, and regulatory agencies will be essential to translate these early successes into standard-of-care treatments that improve patient outcomes in diseases marked by aberrant angiogenesis. The evolving evidence paints a picture of both progress and potential—a future in which VEGF-A–targeted therapies are not only safe and effective but are also tailored precisely to the needs of individual patients, ultimately bridging the gap between bench discoveries and bedside therapies.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.