Small Molecule
C21H23N3O5
783355-60-2
Eftilagimod alfa, also known as efti or IMP321, is a late-stage, first-in-class investigational immunotherapy developed by Immutep Limited. It is a soluble LAG-3Ig fusion protein engineered to function as a potent Antigen-Presenting Cell (APC) activator. Its mechanism of action represents a novel paradigm in immuno-oncology. Unlike antagonistic anti-LAG-3 monoclonal antibodies that block an inhibitory signal on T-cells, eftilagimod alfa acts as a Major Histocompatibility Complex (MHC) Class II agonist, directly stimulating dendritic cells and monocytes. This primary action initiates a broad, systemic immune response that engages both innate and adaptive immunity, culminating in the robust activation and proliferation of CD8+ cytotoxic T-cells.
Clinically, eftilagimod alfa has demonstrated encouraging anti-tumor activity across multiple solid tumors, particularly when used in combination with anti-PD-1 therapy (pembrolizumab). Key indications where it has shown promise include non-small cell lung cancer (NSCLC), head and neck squamous cell carcinoma (HNSCC), and metastatic breast cancer (MBC). A consistent and highly significant finding across its clinical program is the agent's efficacy in patients with low or negative PD-L1 expression—a population with high unmet medical need and often limited benefit from existing checkpoint inhibitors.
The agent is characterized by a consistently favorable and manageable safety profile. The most common adverse events are low-grade, localized injection site reactions. Crucially, the addition of eftilagimod alfa to checkpoint inhibitors or chemotherapy regimens does not appear to introduce significant additive toxicity, making it an attractive combination partner.
Eftilagimod alfa is currently being evaluated in a pivotal Phase III trial (TACTI-004) for first-line NSCLC, representing its most direct path to market. The U.S. Food and Drug Administration (FDA) has granted it Fast Track designation for both first-line NSCLC and first-line HNSCC. The recent successful completion of the FDA's Project Optimus requirements, which formally confirmed the 30 mg optimal biological dose, is a major de-risking milestone that clears a critical path for future Biologics License Application (BLA) submissions and ongoing late-stage development.
Eftilagimod alfa is a complex biologic with multiple identifiers used across scientific literature, clinical trials, and regulatory filings. A consolidated list of its key identifiers is essential for accurate cross-referencing.
Table 1: Eftilagimod alfa - Key Identifiers and Properties
Identifier Type | Value | Source(s) |
---|---|---|
Generic Name | Eftilagimod alfa | 1 |
DrugBank ID | DB16150 | 1 |
CAS Number | 1800476-36-1 | 1 |
UNII | SJ82PK3HWA | 1 |
Common Synonyms | Efti, IMP321, LAG-3Ig, sLAG-3-Ig | 4 |
Development Codes | EOC-202, EDP-202 | 6 |
Investigational Trade Name | ImmuFact | 4 |
Molecular Class | Recombinant Fc Fusion Protein | 5 |
Molecular Weight | ~160 kDa (glycosylated) | 4 |
Route of Administration | Subcutaneous injection | 4 |
Eftilagimod alfa is classified as a large-molecule, recombinant Fc fusion protein, a type of biotech therapeutic.[2] Its molecular architecture is central to its unique immunological function. The molecule is a homodimeric protein with a total molecular weight of approximately 160 kDa.[4] Each monomer consists of the four extracellular domains (D1-D4) of human Lymphocyte Activation Gene-3 (LAG-3) fused to the crystallizable fragment (Fc) region of a human immunoglobulin G1 (IgG1).[4]
This specific design creates a soluble, dimeric version of the LAG-3 protein. The selection of the IgG1 Fc region is a critical engineering choice that extends beyond simply increasing the molecule's half-life. The natural tendency of the IgG1 Fc domains to dimerize is fundamental to the drug's agonistic activity. This dimerization allows eftilagimod alfa to effectively bind to and cross-link multiple MHC Class II molecules on the surface of an APC. This cross-linking event serves as a potent activation signal for the APC, a function that would be significantly less effective with a monomeric protein. Thus, the structural composition is deliberately designed to maximize its function as an MHC Class II agonist.
Detailed characterization of the protein, which is of human origin, has confirmed its complete amino acid sequence, N-linked glycosylation sites, and disulfide bond structure.[10] The estimated molecular formula is , with a calculated average molecular weight of approximately 143 kDa for the non-glycosylated protein backbone.[10]
Eftilagimod alfa is formulated as a solution for subcutaneous (s.c.) injection.[4] This route of administration is strategically aligned with the drug's mechanism of action. The subcutaneous tissue is rich in resident APCs, including dendritic cells and monocytes, which are the primary cellular targets of the drug. Administering eftilagimod alfa directly into this microenvironment ensures high local concentrations, promoting efficient engagement and activation of these key immune cells. Following activation, these APCs migrate to draining lymph nodes to orchestrate a systemic anti-tumor T-cell response. This targeted delivery is mechanistically more direct than intravenous administration and offers the additional benefit of convenience for patients.
Clinical development has explored various dose levels, including 1 mg, 6 mg, and 30 mg, with a 90 mg dose also being evaluated in the ongoing AIPAC-003 trial.[11] Dose-escalation studies demonstrated that higher doses were associated with greater biological activity.[4] Following a comprehensive review of clinical data under the FDA's Project Optimus initiative, the optimal biological dose for ongoing and future studies has been established and agreed upon as 30 mg.[14] The typical dosing schedule in late-stage trials involves a 30 mg subcutaneous injection every two weeks for an initial treatment phase (e.g., the first 6 months or 8 cycles), followed by a maintenance schedule of every three weeks.[16]
Lymphocyte Activation Gene-3 (LAG-3, also known as CD223) is a well-established immune checkpoint receptor. It is primarily expressed on the surface of activated T-cells, including cytotoxic CD8+ T-cells and helper CD4+ T-cells, as well as on regulatory T-cells (Tregs).[1] Its principal function is to act as an inhibitory receptor, or an immune "brake," that negatively regulates T-cell proliferation, activation, and cytokine secretion.[1] The canonical ligand for LAG-3 is MHC Class II, a molecule expressed on the surface of APCs. LAG-3 binds to MHC Class II with a significantly higher affinity than the T-cell co-receptor CD4, allowing it to outcompete CD4 and disrupt the formation of a stable T-cell receptor (TCR) signaling complex. This interaction delivers a potent inhibitory signal into the T-cell, effectively dampening the immune response.[4] Tumors often exploit this pathway to induce T-cell exhaustion and evade immune destruction.
Eftilagimod alfa leverages the LAG-3/MHC Class II interaction in a completely novel and distinct manner. As a soluble, dimeric LAG-3Ig fusion protein, it does not act on T-cells. Instead, it functions as a first-in-class MHC Class II agonist that directly targets APCs.[9] By binding to and cross-linking MHC Class II molecules on the surface of immature dendritic cells and monocytes, eftilagimod alfa provides a powerful activation signal.[4] This interaction mimics the physiological signal that APCs would normally receive from activated T-cells, thereby triggering their maturation and enhancing their capacity to initiate an immune response.[13]
The activation of APCs by eftilagimod alfa sets off a cascade of downstream immunological events that engage both the innate and adaptive immune systems.
The therapeutic approach of eftilagimod alfa is fundamentally different from that of anti-LAG-3 monoclonal antibodies like relatlimab. This distinction can be conceptualized as a "push" versus "pull" mechanism.
This mechanistic distinction has profound clinical implications. The action of eftilagimod alfa is orthogonal and highly complementary to that of PD-1/PD-L1 checkpoint inhibitors. Anti-PD-1 therapies are most effective in immunologically "hot" tumors, which already have a significant T-cell infiltrate but where those T-cells are being suppressed by the PD-1 pathway.[23] Their efficacy is limited in "cold," non-inflamed tumors that lack this pre-existing T-cell presence. Eftilagimod alfa's ability to activate APCs and prime a de novo T-cell response can effectively convert a "cold" tumor microenvironment into a "hot" one. This provides a clear biological rationale for the consistent clinical activity observed in patient populations with low or negative PD-L1 expression in trials such as TACTI-002 and TACTI-003.[9] In this context, eftilagimod alfa acts as an enabling therapy, generating a robust T-cell response that can then be sustained and protected by a co-administered PD-1 inhibitor.
The pharmacokinetic (PK) profile of eftilagimod alfa has been characterized in Phase I clinical trials, most notably the TACTI-mel study in metastatic melanoma.[33] Following subcutaneous administration, the drug exhibits dose-proportional exposure. Analysis across dose levels of 1 mg, 6 mg, and 30 mg demonstrated a corresponding increase in key PK parameters such as maximum plasma concentration () and area under the curve (AUC).[33]
A critical finding from these studies was the establishment of a PK threshold for achieving systemic biological activity. At the 30 mg dose, all patients achieved sustained circulating plasma concentrations of eftilagimod alfa above 1 ng/mL, a level that was not consistently reached at the lower 1 mg and 6 mg doses.[11] This finding was instrumental in the data-driven selection of 30 mg as the optimal biological dose for late-stage development, a decision later affirmed by the FDA.[14] The median time to maximum concentration () following the first administration was approximately 24 hours for the 30 mg dose.[33]
The development of anti-drug antibodies (ADAs) has been observed, with the proportion of ADA-positive patients increasing with higher doses and longer treatment duration. However, the presence of ADAs did not appear to have a negative impact on the drug's pharmacodynamic effects or its clinical efficacy, suggesting they are not neutralizing.[33]
The pharmacodynamic (PD) effects of eftilagimod alfa provide a direct and measurable confirmation of its proposed mechanism of action. Treatment consistently induces a rapid, significant, and sustained increase in the absolute counts of peripheral blood immune cells, including monocytes (the APC precursors) and total lymphocytes.[4]
This is followed by a corresponding increase in the populations of secondary target cells, particularly activated CD4+ helper T-cells and CD8+ cytotoxic T-cells.[11] This cellular response is accompanied by a systemic increase in the plasma levels of Th1-signature biomarkers, most notably IFN-γ and the chemokine CXCL10, which are indicative of a productive anti-tumor immune response.[11]
Crucially, these PD effects have been shown to correlate directly with clinical benefit, providing a powerful validation of the therapeutic hypothesis. In multiple trials, including AIPAC and TACTI-002, early increases in the absolute lymphocyte count (ALC) and the number of circulating activated CD8+ T-cells were significantly associated with improved overall survival.[23] This creates a clear and evidence-based causal chain: the administration of eftilagimod alfa leads to the measurable PD effect of APC and T-cell proliferation, which in turn drives a more effective anti-tumor immune response that translates into the clinical outcome of longer patient survival. This strong PK/PD/efficacy relationship provides a robust biological rationale for the drug's activity and supports the use of these cellular biomarkers for patient monitoring and potentially for patient selection in the future.
The clinical development program for eftilagimod alfa is extensive, spanning multiple solid tumor indications and combination strategies. The program has followed a strategic, data-driven "learn and confirm" approach, where positive signals from early-phase studies have directly informed the design of larger, later-stage trials.
Table 2: Overview of Key Clinical Trials in the Eftilagimod alfa Development Program
Trial Name | NCT ID | Phase | Indication(s) | Combination Therapy | Patient Population | Status |
---|---|---|---|---|---|---|
TACTI-004 | NCT06726265 | III | 1L NSCLC | Efti + Pembrolizumab + Chemo | 1L advanced/metastatic NSCLC, PD-L1 unselected | Recruiting |
TACTI-003 | NCT04811027 | IIb | 1L HNSCC | Efti + Pembrolizumab | 1L recurrent/metastatic HNSCC, by PD-L1 status | Active, not recruiting |
TACTI-002 | NCT03625323 | II | 1L & 2L NSCLC, 2L HNSCC | Efti + Pembrolizumab | PD-L1 unselected NSCLC and HNSCC | Completed |
AIPAC-003 | NCT05747794 | II/III | Metastatic Breast Cancer | Efti + Paclitaxel | HER2-neg/low and TNBC | Recruiting |
AIPAC | NCT02614833 | IIb | Metastatic Breast Cancer | Efti + Paclitaxel | HR+/HER2- MBC post-endocrine therapy | Completed |
INSIGHT-003 | NCT03252938 | I | 1L NSCLC | Efti + Pembrolizumab + Chemo | 1L non-squamous NSCLC | Active, not recruiting |
TACTI-mel | NCT02676869 | I | Metastatic Melanoma | Efti + Pembrolizumab | Unresectable or metastatic melanoma | Completed |
INSIGHT-004 | NCT03252938 | I | Advanced Solid Tumors | Efti + Avelumab | Various advanced solid malignancies | Completed |
NSCLC represents the lead indication for eftilagimod alfa, with an ongoing pivotal Phase III trial.
This open-label, single-arm study evaluated eftilagimod alfa (30 mg s.c.) plus pembrolizumab in a cohort of 114 patients with previously untreated (first-line) metastatic NSCLC, who were unselected for PD-L1 expression.[17] The results were highly encouraging and demonstrated robust anti-tumor activity across all PD-L1 subgroups. The confirmed objective response rate (ORR) was 35.1% (with an unconfirmed ORR of 40.4%), and the disease control rate (DCR) was 72.8%. The combination yielded durable responses, with a median duration of response (DoR) of 21.6 months. Importantly, the median overall survival (OS) was 20.2 months, with 44.7% of patients alive at 24 months, and the median progression-free survival (PFS) was 6.6 months.[9] The activity observed in patients with low (1-49% TPS) and negative (<1% TPS) PD-L1 expression was particularly noteworthy, as these groups represent a high unmet need.[9]
Another cohort within the TACTI-002 trial enrolled 36 patients with metastatic NSCLC who had progressed on or after first-line anti-PD-(L)1-based therapy.[16] In this difficult-to-treat, resistant population, the combination of eftilagimod alfa and pembrolizumab showed signs of overcoming immunotherapy resistance. The confirmed ORR was 8.3% and the DCR was 33.3%. The median OS of 9.9 months was considered promising for this setting. The study suggested that patients with higher PD-L1 expression or those with acquired (as opposed to primary) resistance to prior immunotherapy derived greater benefit.[5]
This investigator-initiated trial was the first to evaluate a triple combination of eftilagimod alfa, pembrolizumab, and standard doublet chemotherapy (carboplatin/pemetrexed) in first-line non-squamous NSCLC.[40] The study reported high response rates, providing a strong rationale for advancing this triple-combination regimen into late-stage testing.[14]
Building on the compelling data from TACTI-002 and INSIGHT-003, Immutep launched the pivotal TACTI-004 trial. This is a large-scale, global, randomized, double-blind study designed to enroll approximately 756 patients with first-line advanced or metastatic NSCLC (both squamous and non-squamous subtypes), irrespective of their PD-L1 status.[18] The trial is comparing the efficacy of eftilagimod alfa plus pembrolizumab and chemotherapy against a control arm of placebo plus pembrolizumab and chemotherapy. The dual primary endpoints are OS and PFS.[18] The trial is actively enrolling patients globally and represents the most direct path to potential regulatory approval for eftilagimod alfa.[15]
HNSCC is another key area of development where eftilagimod alfa has shown significant promise, especially in a patient population with very limited treatment options.
A cohort of 39 patients with second-line HNSCC who had progressed after platinum-based chemotherapy was enrolled in TACTI-002.[17] The final results showed a robust ORR of 29.7%, which included a high rate of complete responses at 13.5%. The median OS was 8.7 months. The benefit was particularly pronounced in patients with high PD-L1 expression (CPS ≥20), where the ORR reached 60% and the median OS was 15.5 months.[13]
This randomized, controlled trial evaluated eftilagimod alfa plus pembrolizumab in first-line HNSCC.[24] The trial's most impactful results came from a single-arm cohort of patients with PD-L1 negative tumors (CPS <1), a group with a very poor prognosis and no effective chemotherapy-free options. In this cohort, the combination achieved a clinically meaningful ORR of 26.9% and a median OS of 17.6 months.[24] This result represents a substantial improvement over historical outcomes for anti-PD-1 monotherapy in this population and has garnered positive feedback from the FDA, opening potential pathways for accelerated approval.[32] The combination also demonstrated improved response rates compared to pembrolizumab monotherapy in the randomized cohorts of PD-L1 positive patients.[24]
The AIPAC trial was a large, randomized, double-blind study that enrolled 227 women with HR-positive, HER2-negative MBC who had progressed on endocrine therapy.[26] It compared eftilagimod alfa plus weekly paclitaxel to placebo plus paclitaxel. The study did not meet its primary endpoint of improving PFS, with a median PFS of approximately 7.3 months in both arms.[26] However, several important secondary and exploratory findings emerged. There was a positive trend toward improved OS in the overall population (20.4 months with efti vs. 17.5 months with placebo; HR 0.88), and this survival benefit became statistically significant in predefined subgroups that align with the drug's mechanism of action (e.g., patients younger than 65, and those with low baseline monocyte counts).[26] Furthermore, eftilagimod alfa treatment led to sustained quality of life, whereas patients in the placebo arm experienced a significant deterioration.[29] The disconnect between the PFS and OS results may suggest that the immune-mediated benefit of an APC activator takes time to manifest, making OS a more relevant long-term endpoint for this class of drugs.
Based on the encouraging OS signal and strong PD data from the AIPAC study, the follow-up AIPAC-003 trial was initiated.[12] This is an integrated Phase II/III study designed to first optimize the dose (30 mg vs. 90 mg) in a Phase II lead-in before proceeding to a large, placebo-controlled Phase III portion powered for OS. The trial is enrolling patients with HER2-negative/low and triple-negative MBC.[12] The safety lead-in portion has been completed, showing that the 90 mg dose is manageable.[50]
Table 3: Summary of Key Efficacy Outcomes for Eftilagimod alfa Combinations Across Major Indications
Trial Cohort | Indication | Combination | ORR (%) | DCR (%) | Median PFS (months) | Median OS (months) |
---|---|---|---|---|---|---|
TACTI-002 | 1L NSCLC (PD-L1 unselected) | Efti + Pembrolizumab | 35.1 | 72.8 | 6.6 | 20.2 |
TACTI-002 | 2L NSCLC (PD-1 refractory) | Efti + Pembrolizumab | 8.3 | 33.3 | 2.1 | 9.9 |
TACTI-003 | 1L HNSCC (CPS <1) | Efti + Pembrolizumab | 26.9 | 58.1 (from cohort B) | N/A | 17.6 |
TACTI-002 | 2L HNSCC (PD-L1 unselected) | Efti + Pembrolizumab | 29.7 | 38.0 | 2.1 | 8.7 |
AIPAC | MBC (HR+/HER2-) | Efti + Paclitaxel | 48.3 | N/A | 7.3 | 20.4 |
Note: Data compiled from multiple sources.[16] ORR and DCR for TACTI-003 are from different cohorts/reports and should be interpreted accordingly.
The breadth of the clinical program highlights a clear strategy to position eftilagimod alfa as a versatile "backbone" immunotherapy. Its consistent safety profile allows for its combination with anti-PD-1 agents, anti-PD-L1 agents, chemotherapy, and even in triple-combination regimens. This adaptability, coupled with its efficacy in diverse tumor types, underscores its potential as a foundational immune-stimulating agent that can be paired with various standards of care to enhance outcomes across the oncology landscape.
A hallmark of eftilagimod alfa across its extensive clinical development program is its favorable and manageable safety profile. This has been consistently demonstrated in combination with checkpoint inhibitors (pembrolizumab, avelumab) and chemotherapy (paclitaxel), as well as in triple-combination regimens.[16]
The most frequently reported treatment-emergent adverse event (TEAE) directly attributable to eftilagimod alfa is low-grade (Grade 1 or 2) injection site reactions, such as erythema or pruritus.[11] Other commonly observed AEs in combination studies, such as fatigue, asthenia, cough, and dyspnea, are generally consistent with the known safety profiles of the partner agents and the underlying disease.[38]
A critical aspect of its safety is the lack of significant additive toxicity. The addition of eftilagimod alfa to existing therapeutic regimens does not appear to introduce novel safety signals or substantially increase the incidence or severity of known toxicities, such as the immune-related adverse events (irAEs) associated with checkpoint inhibitors or the hematological and neurological toxicities of chemotherapy.[24] Rates of treatment discontinuation due to adverse events related to eftilagimod alfa are consistently low across trials.[23] Furthermore, during the dose-escalation phases of development, no dose-limiting toxicities were observed, even at the now-established optimal dose of 30 mg.[11]
This excellent safety profile is a direct consequence of its unique and targeted mechanism of action. By stimulating a specific upstream component of the immune system (APC activation) rather than causing broad, non-specific T-cell disinhibition (as seen with agents like anti-CTLA-4 antibodies), eftilagimod alfa induces a more controlled and physiological immune response. This explains why its primary side effect is a localized reaction at the site of administration—where APCs are being directly activated—and why it does not significantly exacerbate the systemic irAE profile of its combination partners. This favorable safety is a major strategic asset, enabling its evaluation in a wide array of combination strategies that might be prohibitively toxic with other immunotherapy pairings.
Eftilagimod alfa has garnered significant attention from regulatory bodies, reflecting its potential to address areas of high unmet medical need. The U.S. FDA has granted the therapy Fast Track designation for two separate first-line indications: HNSCC and NSCLC.[14] This designation is intended to facilitate the development and expedite the review of drugs that treat serious conditions and fill an unmet medical need.
Furthermore, Immutep has successfully navigated two key FDA programs that have substantially de-risked its late-stage development.
This pattern of deep and positive engagement across multiple special regulatory programs signals strong confidence from the FDA in the drug's clinical data and its potential to offer a meaningful benefit to patients.
Eftilagimod alfa is the lead asset of Immutep Limited (ASX: IMM; NASDAQ: IMMP), a late-stage biotechnology company that is a pioneer in the therapeutic application of the LAG-3 pathway.[4] The cornerstone of the drug's late-stage development is a strategic collaboration with Merck & Co., Inc. (MSD).[5] This partnership is critical for evaluating eftilagimod alfa in combination with Merck's market-leading anti-PD-1 therapy, KEYTRUDA® (pembrolizumab), across the TACTI-002, TACTI-003, and pivotal TACTI-004 trials. This symbiotic relationship provides Immutep with a reliable supply of the standard-of-care combination agent and leverages Merck's vast global infrastructure for conducting large-scale clinical trials, thereby accelerating the path to market. Immutep has also engaged in collaborations with other pharmaceutical companies, such as Merck KGaA and Pfizer for studies involving the anti-PD-L1 antibody avelumab, and with academic centers for investigator-initiated trials.[5]
Eftilagimod alfa is uniquely positioned within the dynamic field of immuno-oncology. It is not merely an addition to the growing class of LAG-3-targeting agents; its distinct agonistic mechanism as an APC activator sets it apart. The ability to generate a de novo immune response, rather than simply reinvigorating a pre-existing one, provides a powerful solution to one of the primary challenges of current checkpoint inhibitor therapy: a lack of efficacy in immunologically "cold" or PD-L1-low tumors.
The path to market appears to be two-fold and strategically sound. The ongoing pivotal TACTI-004 trial in first-line NSCLC represents the most direct route to a broad, blockbuster indication. A positive outcome in this trial has the potential to establish a new global standard of care for the largest oncology market. In parallel, the exceptionally strong data in the niche but high-unmet-need population of first-line HNSCC with CPS <1, supported by the FDA's Project FrontRunner feedback, offers a potential faster, more targeted path to an initial approval.
Looking forward, the potential for eftilagimod alfa is vast. Its excellent safety profile makes it an ideal combination partner, and its broad mechanism of action suggests applicability across a wide range of solid tumors. The ongoing AIPAC-003 trial will be a critical test of its utility in breast cancer, a traditionally less immunogenic tumor type. The robust correlation between pharmacodynamic biomarkers (such as ALC) and clinical outcomes like overall survival provides a clear opportunity for the future development of a companion diagnostic, which could allow for the selection of patients most likely to derive profound and durable benefit, further solidifying the clinical and commercial value of this promising new immunotherapy.
Published at: October 15, 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.