Efdamrofusp alfa, also known by its development code IBI-302, represents a significant evolution in the therapeutic landscape for neovascular retinal diseases. Developed by Innovent Biologics, it is an intravitreally administered, first-in-class bispecific fusion protein engineered to simultaneously address two fundamental pathogenic pathways in neovascular age-related macular degeneration (nAMD) and diabetic macular edema (DME).[1] The core value proposition of Efdamrofusp alfa is built upon a triad of strategic advantages that collectively address the most pressing unmet needs in contemporary retinal care.
First, its novel dual mechanism of action involves the concurrent inhibition of vascular endothelial growth factor (VEGF) and the complement system. By neutralizing both VEGF isoforms and the central complement components C3b and C4b, Efdamrofusp alfa targets both the neovascularization and the chronic inflammation that drive disease progression and vision loss.[3] This integrated approach moves beyond the purely anti-angiogenic focus of legacy therapies.
Second, the clinical development program has provided robust evidence for extended dosing durability. Data from comprehensive Phase 2 trials demonstrate the potential for treatment intervals of 12 weeks or longer (Q12W+) in a majority of patients with nAMD, a feature that directly confronts the high treatment burden associated with the frequent injections required by many current standards of care.[1] This durability is a cornerstone of its clinical and commercial strategy.
Third, and perhaps most consequentially, Efdamrofusp alfa has shown preliminary but highly significant signals of potential disease modification. Emerging data from Phase 2 studies suggest an ability to inhibit the progression of macular atrophy (MA), a primary cause of irreversible vision loss for which no approved preventative treatment currently exists in the nAMD population.[7] This potential anti-atrophic effect could fundamentally alter the long-term goals of therapy from solely managing exudation to preserving retinal structure.
Currently, Efdamrofusp alfa has successfully completed a rigorous Phase 2 program in nAMD, establishing non-inferiority in visual acuity gains compared to the standard-of-care, aflibercept, while demonstrating superior anatomical outcomes and durability.[6] It has advanced into a pivotal Phase 3 registration trial (STAR) for nAMD and is concurrently being evaluated in a Phase 2 trial for DME.[1] Pending the successful outcome of these late-stage trials, Efdamrofusp alfa is poised to emerge as a formidable new therapeutic option, offering a highly differentiated profile that could challenge and redefine the standard of care for millions of patients with retinal disease.
Efdamrofusp alfa is a meticulously engineered, recombinant, fully human bispecific fusion protein with an estimated molecular weight of 140.4 kDa.[3] Its sophisticated architecture is designed for high-affinity, bivalent targeting and enhanced stability within the vitreous humor. The molecule's structure consists of two functionally distinct domains fused to the Fc fragment of human immunoglobulin G1 (IgG1).[3]
The formal chemical name provides a precise description of its composition: a fusion protein incorporating the domain 2 fragment of FLT1 (fms-related tyrosine kinase 1, also known as VEGFR-1) and the domain 3 fragment of KDR (kinase insert domain receptor, or VEGFR-2), which are then fused to the human IGHG1 Fc region.[3] This design creates a potent "VEGF trap" at one end of the molecule and a novel complement-binding domain at the other. Its estimated molecular formula is
C6240H9740O1872N1700S58.[12] For clinical use, Efdamrofusp alfa is supplied as a sterile, colorless to light yellow liquid intended for intravitreal administration, identified by the Chemical Abstracts Service (CAS) Number 2375661-82-6.[3]
The therapeutic innovation of Efdamrofusp alfa lies in its ability to engage two distinct, yet interconnected, pathological axes of retinal disease with a single molecular entity. This dual-targeting strategy represents a calculated departure from the single-pathway inhibition that has defined retinal pharmacotherapy for over a decade. The pathogenesis of diseases like nAMD and DME is understood to be multifactorial, involving a complex interplay between VEGF-driven angiogenesis and complement-mediated inflammation.[4] Standard anti-VEGF monotherapies have proven highly effective at controlling the former but do not address the latter, creating a therapeutic ceiling that manifests as treatment resistance, long-term efficacy attenuation, and the inexorable progression of atrophic changes in some patients.[1] By designing a molecule that incorporates a complement inhibitor, the developers of Efdamrofusp alfa have sought to address both pathways simultaneously. This synergistic approach is hypothesized not only to provide more comprehensive control of neovascularization and edema but also to quell the chronic, low-grade inflammation that contributes to photoreceptor and RPE cell death over time. This represents a fundamental shift in therapeutic philosophy—from a palliative, anti-exudative treatment to one that is potentially disease-modifying.
The N-terminal of the Efdamrofusp alfa protein functions as a high-affinity VEGF-binding domain.[5] This portion of the molecule acts as a potent decoy receptor or "VEGF trap," a well-validated mechanism for achieving anti-angiogenic effects in the eye.
The C-terminal domain of Efdamrofusp alfa confers its unique anti-inflammatory properties and represents its primary point of differentiation from other advanced retinal therapies.[5] This domain is designed to intervene at a crucial juncture of the complement cascade, a key component of the innate immune system that has been strongly implicated in the pathogenesis of AMD.
The therapeutic rationale for Efdamrofusp alfa was rigorously tested and validated in a series of preclinical studies that provided strong proof-of-concept for its dual mechanism of action and established a clear pathway toward clinical development.
These preclinical studies provided the first critical evidence linking the dual-target mechanism to a potentially disease-modifying outcome. A particularly profound finding from the mouse model was that the dual inhibition of VEGF and complement "was found to further inhibit macrophage infiltration and M2 macrophage polarization".[4] This observation was highly significant. Macrophages are key inflammatory cells deeply involved in the pathogenesis and progression of AMD, with M2-polarized macrophages specifically implicated in processes of fibrosis and tissue remodeling that lead to permanent structural damage. The ability of Efdamrofusp alfa to modulate this specific immune cell activity provided a direct biological rationale for its potential to inhibit the development of fibrosis and macular atrophy in patients. This finding elevated the drug's proposed mechanism beyond simple protein neutralization to the active modulation of the cellular immune response within the retina. This preclinical observation became the foundational hypothesis for the later, successful exploration of macular atrophy as a key secondary endpoint in human clinical trials, where preliminary positive signals were ultimately observed.[1]
Building on the strong preclinical foundation, Innovent Biologics initiated a comprehensive clinical development program to evaluate the safety, efficacy, and durability of Efdamrofusp alfa in patients with nAMD.
The first-in-human evaluation of Efdamrofusp alfa was conducted in a Phase 1b study designed primarily to assess its safety and tolerability in the target patient population.[4] The trial was a prospective, randomized, open-label, multiple ascending-dose study conducted in China, enrolling 18 patients with active nAMD. Participants were randomized in a 2:1 ratio to receive intravitreal injections of either Efdamrofusp alfa (initially 2 mg, then escalated to 4 mg) or the active comparator, aflibercept 2 mg.[4]
The study successfully met its primary objectives, establishing a favorable safety profile for Efdamrofusp alfa. No dose-limiting toxicities or ocular serious adverse events were reported during the study period.[4] All observed ocular treatment-emergent adverse events (TEAEs) were characterized as mild-to-moderate in severity and were deemed to be related to the intravitreal injection procedure itself, rather than the drug substance.[4] The drug was well-tolerated at both dose levels, providing the necessary safety assurance to proceed to larger, more definitive Phase 2 trials.[4] In addition to the positive safety findings, promising efficacy signals were observed, with patients in the Efdamrofusp alfa arms showing clinically meaningful improvements in Best-Corrected Visual Acuity (BCVA) and reductions in Central Subfield Thickness (CST) as measured by optical coherence tomography (OCT).[4]
The Phase 2 program for Efdamrofusp alfa was robust, comprising two separate studies that enrolled a combined total of over 360 participants with nAMD. This provided a substantial dataset to characterize the drug's efficacy and durability ahead of Phase 3.[1] The cornerstone of this program was a randomized, double-masked, active-controlled trial (NCT05403749) that enrolled 132 Chinese patients with active CNV. This study was designed to compare two high-dose formulations of Efdamrofusp alfa (6.4 mg and 8.0 mg) against the standard-of-care, aflibercept 2.0 mg.[6]
The trial successfully met its primary endpoint, which was the mean change in BCVA from baseline to week 40. Both the 6.4 mg and 8.0 mg doses of Efdamrofusp alfa demonstrated non-inferior BCVA gains compared to the aflibercept 2.0 mg arm.[6]
In terms of anatomical outcomes, Efdamrofusp alfa demonstrated robust and numerically superior reductions in retinal fluid as measured by CST, indicating more profound retinal drying compared to aflibercept.
A pivotal finding of the Phase 2 program was the confirmation of the drug's potential for extended-interval dosing. After receiving initial monthly loading doses, a high proportion of patients in the Efdamrofusp alfa arms were able to maintain their visual and anatomical gains while on a 12-week (Q12W) dosing interval.
The key efficacy outcomes from the Phase 2 trial (NCT05403749) are summarized below.
Endpoint | Efdamrofusp Alfa (6.4 mg) | Efdamrofusp Alfa (8.0 mg) | Aflibercept (2.0 mg) |
---|---|---|---|
Mean Change in BCVA (letters) at Week 52 | +10.8 | +11.3 | +10.0 |
Mean Change in CST (µm) at Week 52 | -154.6 | -174.7 | -131.2 |
% Patients on Q12W Dosing | ~81% | ~88% | N/A (Fixed Q8W) |
Based on the strength of the Phase 2 data, Innovent initiated the pivotal STAR study in October 2023. This is a large-scale, randomized, double-masked, active-controlled Phase 3 clinical trial designed to provide the definitive evidence required to support a new drug application for Efdamrofusp alfa in the treatment of nAMD.[1]
The design of the STAR trial is noteworthy for its strategic foresight. Rather than conducting a simple non-inferiority trial with fixed dosing schedules, Innovent has implemented a PTI regimen for the Efdamrofusp alfa arm. This approach closely mimics the "treat-and-extend" (T&E) protocols that have become the standard of care in modern clinical practice, where retina specialists individualize treatment frequency based on patient response. By allowing the dosing interval for Efdamrofusp alfa to be extended up to Q16W based on disease activity, while holding the aflibercept arm to a standard Q8W schedule, the trial is explicitly designed to generate definitive evidence of a reduced treatment burden. This design demonstrates a high degree of confidence from the developer in the drug's durability and aims to produce data that will be immediately relevant and persuasive to practicing clinicians, moving beyond a simple demonstration of non-inferiority on a single visual acuity endpoint to prove superiority in real-world applicability.
The strong scientific rationale for Efdamrofusp alfa's dual mechanism extends logically to the treatment of diabetic macular edema. The pathophysiology of DME, much like nAMD, is driven by a combination of VEGF-mediated increases in vascular permeability and a significant, chronic inflammatory component that contributes to the breakdown of the blood-retinal barrier.[5] This makes DME an ideal second indication for a therapeutic agent that can simultaneously target both angiogenesis and complement-mediated inflammation.
The most strategically significant element of the DME clinical program is the choice of the active comparator: faricimab (Vabysmo).[5] This decision represents a bold and confident "bispecific vs. bispecific" clinical showdown. Faricimab is itself a bispecific antibody, targeting VEGF-A and Angiopoietin-2 (Ang-2), a separate pathway involved in vascular stability and inflammation.[20] It is the newest and most advanced dual-target therapy currently on the market and is considered the benchmark for efficacy and durability in DME.
By choosing faricimab as the comparator, Innovent is deliberately bypassing a comparison against an older standard of care like aflibercept. This is a high-risk, high-reward strategy. A successful outcome, demonstrating non-inferiority or superiority to faricimab, would immediately position Efdamrofusp alfa not merely as another alternative, but as a leading therapeutic option at the forefront of the market. This trial design signals the company's strong belief that targeting the VEGF/Complement axis may offer distinct advantages over targeting the VEGF/Ang-2 axis in the context of DME. It is a clear declaration of intent to compete at the highest level of the market, seeking to establish a new standard of care based on a mechanistically distinct approach to dual-pathway inhibition.
Across the entirety of its clinical development program to date, encompassing Phase 1 and Phase 2 trials in over 360 patients with nAMD and DME, Efdamrofusp alfa has consistently demonstrated a favorable and manageable safety profile. The overall safety findings have been repeatedly described as comparable to those of established anti-VEGF agents, such as aflibercept, with no new or unexpected safety signals identified.[1]
In the development of novel intraocular therapies, a clean and predictable safety profile is of paramount importance, often weighing as heavily as efficacy in clinical decision-making. The ophthalmology community is highly sensitive to the risk of drug-induced intraocular inflammation, as the uptake of previous novel anti-VEGF agents has been significantly hampered by concerns over rare but severe inflammatory side effects. The fact that the safety profile of Efdamrofusp alfa is consistently described as "similar to aflibercept" and, crucially, lacks specific concerning signals like retinal vasculitis, is a major asset. For practicing clinicians, a predictable safety profile that aligns with their decades of experience managing patients on anti-VEGF therapy would substantially lower the barrier to adoption, provided that the efficacy and durability are confirmed in Phase 3. In this context, the absence of new safety concerns is not merely a footnote but a critical competitive advantage.
The most compelling and potentially paradigm-shifting data to emerge from the Efdamrofusp alfa clinical program is the preliminary evidence suggesting an ability to inhibit the development and progression of macular atrophy (MA). This finding was highlighted by Professor Xiaodong Sun, a principal investigator for the clinical trials, who noted that "preliminary positive signals have also been observed in the inhibition of fibrosis and macular atrophy," instilling great confidence in the research community.[1]
This observed reduction in macular atrophy is not an incidental or unexpected finding; rather, it appears to be a direct therapeutic consequence of the drug's unique anti-complement mechanism of action. There is a clear and logical line of evidence supporting this hypothesis, stretching from foundational science through preclinical models to the clinical data. The complement system has been strongly and unequivocally implicated in the pathogenesis of dry AMD and the development of geographic atrophy.[4] This link is so well-established that the first drugs approved for the treatment of GA, such as avacincaptad pegol, are inhibitors of the complement cascade.[20] Efdamrofusp alfa is the first therapeutic agent to integrate this validated anti-complement mechanism into the treatment paradigm for
neovascular AMD. Furthermore, the preclinical studies provided a cellular basis for this effect, showing that Efdamrofusp alfa could modulate macrophage activity, a key cellular process involved in the development of atrophy.[4] Therefore, the clinical signal of reduced MA can be interpreted as the logical and intended therapeutic outcome of targeting a known driver of retinal atrophy. If this effect is confirmed in the ongoing Phase 3 STAR trial, Efdamrofusp alfa would become the first drug capable of simultaneously controlling neovascularization and protecting against the long-term atrophic consequences of the disease, fundamentally changing the goals and expectations of nAMD therapy.
Efdamrofusp alfa is entering a dynamic and competitive market for retinal disease therapies. Its positioning will be defined by its performance relative to both established and new standards of care.
As articulated by clinical experts involved in the trials, current anti-VEGF therapy, while revolutionary, is constrained by significant limitations. These include the substantial treatment burden imposed by frequent injections, the phenomenon of long-term efficacy attenuation, and, most critically, the inability to prevent the progression of underlying atrophic and fibrotic processes that lead to permanent vision loss.[1]
Efdamrofusp alfa has been specifically engineered to address these unmet needs. Its profile—combining extended dosing durability, robust efficacy in improving vision and drying the retina, a favorable safety profile, and its unique potential for anti-atrophic and anti-fibrotic effects—positions it as a highly promising solution to the current challenges in retinal care.[1]
In conclusion, Efdamrofusp alfa stands out as a highly innovative and scientifically rational therapeutic candidate. Its dual mechanism is supported by a strong and coherent body of evidence from preclinical studies through mid-stage clinical trials. The success of the ongoing Phase 3 STAR trial will be the final determinant of its future role. Confirmation of its durability and, most importantly, its benefit in reducing macular atrophy would solidify its position as a potential future standard of care for major retinal diseases. The development of Efdamrofusp alfa, alongside other advanced therapies, signals the dawn of a new era in ophthalmology—one that is moving beyond simple anti-angiogenesis toward more comprehensive, durable, and truly disease-modifying treatments.
Published at: September 9, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.