What is the mechanism of action of Odronextamab?

7 March 2025

Introduction to Odronextamab 
 
Odronextamab is a fully-human, IgG4-based bispecific antibody designed with a unique mechanism that bridges innate and adaptive immunity to provide targeted antitumor activity. It has been engineered to simultaneously bind to two distinct antigens: CD20 expressed on B-cell malignancies and CD3 present on T cells. This design not only facilitates the physical proximity of T cells to the malignant B cells but also activates the T cells directly, triggering potent antitumor responses. Its classification as a bispecific T-cell engager (BiTE) sets it apart from conventional monoclonal antibodies, as it combines the specificity of monoclonal antibody targeting with the immune cell‐redirecting capabilities inherent in bispecific constructs. Odronextamab’s development has evolved from an extensive preclinical research phase that established the translational value of dose‐escalation strategies to robust clinical trials that continue to assess its efficacy and safety profile. 

Clinical Indications 
Clinically, odronextamab is primarily investigated in the realm of B-cell malignancies. It is particularly aimed at treating patients with relapsed or refractory forms of Non-Hodgkin lymphoma (NHL), including diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL). These malignancies often exhibit resistance to conventional treatments and even to CD20-directed monotherapies, which underscores the need for innovative therapeutic approaches. By leveraging the dual-target binding property, odronextamab offers an alternative mechanism by which the immune system can be harnessed to effect tumor cell killing in patients who have shown poor responses to previous therapies, including those already exposed to chimeric antigen receptor (CAR) T-cell therapy. 

Biological Mechanism of Action 
Molecular Targets 
At the molecular level, odronextamab functions by recognizing and binding two distinct antigens: CD20 and CD3. The CD20 antigen is a well-characterized B-cell surface molecule expressed on nearly all B-cell malignancies. Binding to CD20 allows the antibody to anchor onto the tumor cell, effectively marking it for immune destruction. The other binding site is directed towards CD3—a component of the T-cell receptor complex present on T cells. This simultaneous binding results in a physical “bridge” that brings T cells into close proximity with CD20-positive B cells. 

This dual specificity is critical for several reasons. First, by binding to CD20, odronextamab ensures that its cytotoxic activity is primarily directed towards malignant B cells while sparing non-target cells that do not express the antigen. Second, engagement with CD3 triggers an activation cascade within the T cells, independent of traditional T-cell receptor (TCR) recognition of antigen-loaded major histocompatibility complexes (MHC). This property is particularly advantageous in immunologically “cold” tumors where conventional antigen presentation is impaired or down-regulated, thus overcoming one of the significant hurdles associated with T-cell mediated immune responses. 

Furthermore, the engagement of CD3 on T cells not only promotes T-cell activation but also initiates intracellular signaling cascades that lead to the release of cytokines and cytolytic granules. These granules, which contain perforin and granzymes, are instrumental in inducing apoptosis in the target B cells. In summary, the molecular interplay involves a “two-hit” strategy: first, tagging the malignant cell via CD20 binding and second, activating T cells via CD3 engagement, which ultimately results in a robust, localized cytotoxic response. 

Cellular Pathways and Interactions 
Once odronextamab binds to both CD20 on the tumor cell and CD3 on the T cell, it establishes an immunological synapse—a specialized junction that mimics the natural interactions between antigen-presenting cells and T lymphocytes. This synapse allows the T cells to receive activation signals even in the absence of classical antigen recognition through the TCR. The ensuing T-cell activation results in a cascade of intracellular events, including a rapid increase in calcium flux, activation of protein kinase C, and mobilization of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). These signaling events collectively lead to T-cell proliferation, differentiation, and the secretion of pro-inflammatory cytokines such as interleukin-2 (IL-2) and interferon-gamma (IFN-γ). 

The binding of odronextamab to CD3 essentially bypasses the need for co-stimulation through engagement of conventional major histocompatibility complex (MHC)-peptide complexes, thereby providing a more direct and potent route to T-cell activation. This is particularly relevant in malignancies where tumor cells evade immune detection by down-regulating MHC molecules. Furthermore, the T-cell activation induced by odronextamab engages cytotoxic mechanisms that lead to antibody-dependent cellular cytotoxicity (ADCC) and direct T-cell-mediated killing. 

In addition to initiating direct cytotoxic responses, the cross-linking of T cells and malignant cells facilitates the production of cytokines that augment the local immune environment. This cytokine release not only enhances the recruitment of other immune cells but also modifies the tumor microenvironment, potentially shifting it from an immunosuppressive to a pro-inflammatory state. Such modulation of the tumor microenvironment represents an emergent property of bispecific antibody therapy that complements its direct cytolytic effects. 

Pharmacodynamics and Pharmacokinetics 
Absorption and Distribution 
The pharmacokinetic behavior of odronextamab is carefully designed to maximize its therapeutic window and ensure that effective concentrations are sustained in the bloodstream. Preclinical and early-phase clinical studies have provided insights into its absorption and distribution profiles. When administered intravenously, odronextamab exhibits a distribution pattern that allows it to effectively reach both peripheral blood compartments and lymphoid tissues where B-cell malignancies commonly reside. The IgG4 backbone of odronextamab typically contributes to a prolonged half-life, thereby supporting sustained exposure that is critical for continuous T-cell engagement and tumor cell targeting. 

During preclinical studies, pharmacokinetic data demonstrated that serum concentration levels required to achieve cytokine release in vitro corresponded closely with those observed in the clinical setting following dose escalation. This correlation was instrumental in guiding dose selection and escalation parameters in first‐in‐human studies, ensuring that therapeutic levels are achieved while mitigating the risk of excessive immune activation. The widespread tissue distribution and relatively predictable pharmacokinetic profile of odronextamab underpin its potential for use as an off‐the‐shelf therapy, offering practical advantages over autologous treatments that require patient-specific manufacturing processes. 

Metabolism and Excretion 
Like other monoclonal antibodies and biologics, odronextamab undergoes catabolic degradation rather than being extensively metabolized through hepatic enzymes. This degradation involves intracellular lysosomal proteolysis following cellular uptake via the neonatal Fc receptor (FcRn) recycling pathway—a mechanism that protects IgG antibodies from rapid clearance. Thus, the elimination of odronextamab occurs predominantly via proteolytic catabolism into peptides and amino acids. 

Furthermore, the design of odronextamab minimizes interactions with cytochrome P450 enzyme systems, thereby reducing the likelihood of drug-drug interactions commonly seen with small-molecule therapeutics. This property is particularly beneficial in polytherapy settings where patients may be receiving multiple agents concurrently. In addition, this metabolic pathway contributes to a predictable safety profile, with minimal concerns regarding accumulation or toxicity from metabolic intermediates. 

This robust pharmacokinetic foundation is essential for maintaining consistent drug exposure over prolonged treatment periods and for optimizing immunogenic responses. Effective clearance mechanisms coupled with long half-life permit dosing flexibility, including step-up regimens that can be tailored to individual patient tolerability and disease burden. The impact of these pharmacokinetic features has been a cornerstone in the design of clinical trials and is continuously monitored throughout the clinical development program of odronextamab. 

Clinical Implications and Research 
Efficacy in Clinical Trials 
Clinically, odronextamab has demonstrated promising efficacy in early-phase trials involving patients with relapsed or refractory B-cell malignancies. In these studies, the bispecific mechanism of action has translated into substantial objective response rates (ORR) and complete responses (CR) in a subset of patients. For example, the Phase 1 expansion cohort reported ORRs that ranged from 48% to 52% in patients, including those who had previously failed CAR T-cell therapy—a patient population with historically poor outcomes. 

The clinical efficacy of odronextamab is particularly notable for its ability to generate deep and durable responses in high-risk patient subgroups, such as those with high International Prognostic Index (IPI) scores or aggressive disease phenotypes. The robust engagement of T cells, as evidenced by cytokine release and the direct cytotoxic effects observed in clinical samples, underscores the translational success of the immunological synapse formation mediated by the antibody. 

Furthermore, the engagement of the patient’s own immune system via a bispecific approach represents a departure from conventional chemotherapy and targeted therapies that may rely on a single mechanism. The cumulative clinical data not only highlights the activity of odronextamab in controlling tumor growth but also confirms the feasibility of its dosing strategy, which leverages step-up dosing to mitigate the early onset of cytokine release syndrome (CRS) while achieving sustained therapeutic levels. 

Safety and Side Effects 
Alongside its promising efficacy, odronextamab’s safety profile is of paramount importance. Clinical studies have observed that adverse events are generally manageable and consistent with those expected from immune-engaging therapies. One of the most frequently reported adverse events is cytokine release syndrome (CRS), which is an expected pharmacodynamic effect given the robust T-cell activation elicited by odronextamab. 

CRS, when it occurs, is usually of low grade and can be effectively managed with supportive care and step-up dosing schedules designed to gradually acclimate the patient’s immune system to the active agent. In the clinical trial data, the incidence of Grade 3 CRS events was low, and the majority of events resolved within a few days of onset, demonstrating that the dosing regimen is appropriately calibrated to balance efficacy and safety. 

Moreover, the side effect profile of odronextamab generally compares favorably to that of conventional chemotherapeutic agents, with fewer systemic toxicities and a lower risk of long-term adverse effects. Its targeted mechanism allows for preferential cytotoxicity against cancerous cells, minimizing collateral damage to healthy tissues. The overall tolerability observed in clinical trials supports its future development both as a monotherapy and potentially in combination with other therapeutic modalities. 

Future Research Directions 
Ongoing Studies 
The research on odronextamab remains dynamic and forward-looking. Numerous ongoing clinical trials continue to refine our understanding of its therapeutic window, optimal dosing strategies, and long-term efficacy. A pivotal Phase 2 trial is underway, expanding odronextamab’s evaluation in DLBCL and FL, and additional Phase 3 studies are in the planning stages to compare its efficacy against standard-of-care regimens in a randomized setting. 

Moreover, translational studies are actively investigating the predictive value of circulating tumor DNA (ctDNA) as a biomarker for response to odronextamab treatment. These studies aim to provide real-time insights into tumor dynamics and potentially enable early intervention strategies for patients showing signs of treatment resistance or relapse. The robust clinical research infrastructure built around odronextamab not only enhances our understanding of its mechanism but also paves the way for regulatory approvals in multiple regions, including the European Union and the United States. 

In addition, ongoing post-marketing surveillance and expanded safety monitoring are designed to detect any emerging safety signals over longer periods of administration. This continued research effort underscores the commitment to ensuring that the benefits of odronextamab, delivered through its novel mechanistic approach, are both maximized and sustained in diverse patient populations. 

Potential for Combination Therapies 
Another promising avenue for the future lies in the combination of odronextamab with other therapeutic agents. The bispecific nature of odronextamab aligns well with combination strategies where complementary mechanisms of action can lead to enhanced efficacy. For instance, combining odronextamab with agents that provide additional costimulatory signals such as CD28-targeted therapies might further augment T-cell activation and overcome any intrinsic resistance associated with monotherapy. 

Preclinical studies have demonstrated that combining odronextamab with other immunomodulatory therapies can enhance antitumor immunity. For example, an innovative approach involved pairing odronextamab with a CD22-targeted CD28 bispecific antibody, which significantly increased T-cell activation and induced more profound tumor cell apoptosis in refractory DLBCL models. Such combinations could produce synergistic effects that not only potentiate tumor cell killing but also alter the tumor microenvironment to favor sustained immune surveillance and longer-term remissions. 

Furthermore, combining odronextamab with other standard cytotoxic or targeted therapies may improve clinical outcomes by exploiting different mechanisms of action. This combinatorial approach is particularly valuable in heavily pretreated or high-risk patient populations where monotherapy might be insufficient. Ongoing and planned clinical trials are expected to explore combinations with chemotherapy, checkpoint inhibitors, and other novel agents, with early data suggesting acceptable tolerability and promising efficacy. 

These combination strategies also open up opportunities to address potential limitations, such as transient immunosuppression or antigen escape, by providing a multi-pronged attack on tumor cells. Importantly, the flexibility of odronextamab’s mechanism allows it to be integrated into multiple treatment paradigms, potentially serving as both frontline therapy when combined with conventional agents or as a salvage therapy in patients who have relapsed from previous treatments. 

Detailed Conclusion 
Odronextamab represents a significant advancement in the therapeutic realm of immuno-oncology as a bispecific antibody that harnesses the power of the patient’s own immune system. Its mechanism of action is rooted in its dual binding capacity: targeting CD20 on malignant B cells and engaging CD3 on cytotoxic T cells. By physically bridging these two cell types, odronextamab induces the formation of an immunological synapse that bypasses the classical antigen recognition pathways, leading to rapid T-cell activation and targeted tumor cell lysis. This unique mode of action not only facilitates direct cytotoxic effects against tumor cells but also modulates the tumor microenvironment to favor prolonged antitumor responses. 

From a pharmacokinetic and pharmacodynamic perspective, odronextamab's design ensures robust tissue distribution, sustained serum concentrations, and predictable metabolic clearance via proteolytic pathways. These properties allow for an effective step-up dosing regimen that minimizes the risks associated with cytokine release syndrome while maintaining therapeutic efficacy. Toxicity profiles emerging from clinical trials have been acceptable, with manageable side effects that reinforce its distinguishing advantage over many conventional chemotherapies. 

Clinically, early-phase trials have demonstrated substantial efficacy in challenging patient populations, including those with relapsed or refractory DLBCL and FL. The substantial objective response and complete response rates observed have positioned odronextamab as a promising candidate for further clinical development. Such efficacy is particularly noteworthy given its ability to induce deep, lasting responses in patients who previously failed other immune-based therapies such as CAR T-cell therapy. 

Looking toward the future, ongoing studies are refining both the safety and efficacy parameters of odronextamab, while also exploring its integration into combination therapy regimens. Whether combined with additional immunomodulatory agents, costimulatory antibodies, or cytotoxic drugs, the future of odronextamab research is set to uncover new therapeutic niches and potentially broaden its application across various B-cell malignancies. These clinical investigations, along with translational research on biomarkers like ctDNA, promise to deepen our understanding of how best to deploy this innovative therapy to maximize patient outcomes. 

In conclusion, odronextamab’s mechanism of action, marked by its dual-specific binding and resultant T-cell activation, provides a paradigm shift in cancer immunotherapy. Its ability to directly engage the immune system against tumor cells, combined with a favorable pharmacokinetic profile and an emerging safety record, underscores its potential as an effective treatment for relapsed or refractory B-cell malignancies. The ongoing rigorous clinical and preclinical research efforts continue to emphasize its promise, paving the way for future innovations that may combine odronextamab with other therapeutic modalities to achieve even greater clinical success. This comprehensive understanding of its mechanism of action not only supports its continued clinical development but also provides critical insights that could inform the design of next-generation bispecific antibodies in the evolving field of immuno-oncology.

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.