What is the therapeutic class of Elranatamab?

7 March 2025
Introduction to Elranatamab
Elranatamab is an innovative therapeutic agent that has emerged as a promising immunotherapy for multiple myeloma. It belongs to the class of bispecific antibodies, and more specifically, it is a BCMA-CD3 targeting bispecific T-cell engager. Over the past few years, extensive preclinical and clinical studies have paved the way for its accelerated development and regulatory review for the treatment of relapsed or refractory multiple myeloma. The clinical data, which are primarily derived from well-structured studies such as the Phase 2 MagnetisMM-3 trial, have underlined its potent anti-myeloma activity and a manageable safety profile.

Basic Information and Development
Elranatamab, sometimes referred to as elranatamab-bcmm or marketed under the brand name ELREXFIO™, is an investigational agent developed by Pfizer. It is designed as a bispecific antibody that simultaneously binds to B-cell maturation antigen (BCMA) expressed on multiple myeloma cells and CD3 on T cells. By bridging these two cell types, elranatamab directs the cytotoxic potential of T cells to malignant plasma cells, thus inducing T cell-mediated killing of myeloma cells. Its development process has involved an array of preclinical experiments assessing its binding affinity, immune-stimulatory capacity, and preliminary safety, as well as clinical trials that have investigated multiple dosing regimens and the mitigation of adverse events such as cytokine release syndrome (CRS). The compound is administered via subcutaneous injection, a route which offers potential benefits in terms of improved pharmacokinetics and tolerability when compared to intravenous infusions.

Overview of Elranatamab in Medicine
In the context of medicine, elranatamab represents a major advancement in the immunotherapy of multiple myeloma. It is categorized as a bispecific T-cell engager (BiTE)-like antibody, which reflects its mechanism of action and its therapeutic application. With multiple myeloma remaining an incurable but treatable plasma cell malignancy, the development of new modalities that can overcome refractory disease and provide deeper and more durable responses is a significant breakthrough. The clinical application of elranatamab has been driven by the high unmet need in patients who have been heavily pretreated with therapies including proteasome inhibitors, immunomodulatory agents, and monoclonal antibodies targeting CD38. Its role, therefore, is not just limited to being another anti-myeloma drug but also as an innovative platform in cancer immunotherapy that manipulates T-cell activity to achieve cytotoxicity against malignant cells.

Therapeutic Classification of Elranatamab
Elranatamab is firmly placed within the therapeutic class of bispecific antibodies. It is specifically designed as a T-cell engaging antibody that redirects the immune system towards malignant plasma cells. This therapeutic approach leverages the specificity of monoclonal antibodies while recruiting the inherent cytotoxic power of T cells—forming a bridge between effector and target cells.

Mechanism of Action
The mechanism of action of elranatamab is based on its dual-targeting property. One arm of the antibody binds to BCMA, a protein overexpressed on the surface of multiple myeloma cells, while the other arm attaches to CD3, which is a component of the T-cell receptor complex found on T cells. This simultaneous binding has a significant impact:

• By binding to BCMA, elranatamab distinguishes malignant plasma cells from healthy cells. BCMA is an attractive target as it is almost exclusively expressed on plasma cells and thus limits off-target toxicity.

• When the CD3 arm of elranatamab binds to T cells, it triggers T-cell activation. Once activated, T cells release cytotoxic granules and cytokines that lead to the destruction of the bound myeloma cells. This T cell-mediated cytotoxicity is the cornerstone of its anti-tumor activity.

Furthermore, the engineering of the binding affinity for both BCMA and CD3 has been optimized to maximize therapeutic efficacy while reducing the likelihood of overstimulation that can lead to adverse events like CRS. This intricate balance between effective immune activation and toxicity mitigation has been a critical focus during its development. The ability to activate T cells in a controlled and potent manner is what classifies elranatamab as a bispecific T-cell engager, and it aligns with similar molecules designed to bring tumor cells into close proximity with cytotoxic lymphocytes.

Comparison with Similar Therapeutics
When comparing elranatamab with other therapeutics, the similarities and differences become apparent in both structure and clinical application. Like other bispecific antibodies such as teclistamab and blinatumomab, elranatamab is designed to harness T-cell mediated immunity. However, whereas blinatumomab has been established in the treatment of relapsed or refractory B-cell acute lymphoblastic leukemia (ALL) by targeting CD19 and CD3, elranatamab focuses on BCMA in multiple myeloma, addressing a distinct and challenging malignancy.

• Teclistamab is another anti-BCMA bispecific antibody and shares the common mechanism of bridging T cells to myeloma cells. However, differences in dosing regimen (weekly versus biweekly), adverse event profiles, and pharmacokinetics have provided clinicians with alternative treatment options that may be better suited to individual patient needs. For instance, elranatamab’s subcutaneous route and engineered dosing strategy (with step-up priming doses) are designed to reduce CRS severity and improve patient convenience.

• Blinatumomab, although similar in its bispecific T-cell engaging nature, is constructed as a small, single-chain molecule that lacks an Fc region, leading to a shorter half-life necessitating continuous infusion. In contrast, elranatamab is a full-length bispecific antibody which offers a longer half-life and thus the potential for less frequent dosing, a key practical difference in terms of patient management.

In summary, elranatamab is comparable to other bispecific T-cell engagers but distinguishes itself through its target (BCMA), method of administration (subcutaneous injection), and optimized pharmacokinetic and safety profile, which positions it uniquely within the therapeutic arsenal for multiple myeloma treatment.

Clinical Applications
Elranatamab’s use is mainly centered on treating relapsed or refractory multiple myeloma, particularly in patients who have exhausted other treatment options. As a bispecific antibody bridging T cells and myeloma cells, its therapeutic value lies in leveraging the immune system’s ability to selectively target and eliminate malignant cells.

Current and Potential Uses
Currently, elranatamab has received accelerated regulatory attention following promising data from clinical trials. It has been granted accelerated approval under conditions that may allow continuation of its use pending confirmatory clinical trials demonstrating long-term benefit. Its primary indication is for patients with relapsed or refractory multiple myeloma who have undergone several prior lines of therapy, including proteasome inhibitors, immunomodulatory drugs (IMiDs), and anti-CD38 monoclonal antibodies. The targeting of BCMA, which is abundantly expressed on multiple myeloma cells, opens a novel path especially in cases deemed “triple-class refractory,” where available treatment options are severely limited.

Potentially, elranatamab may also be used in combination with other immunotherapeutic agents or standard treatments to enhance anti-tumor effects. For example, there are ongoing trials investigating combinations with daratumumab (an anti-CD38 monoclonal antibody), proteasome inhibitors, and other novel agents to explore synergistic effects and further improve response rates. Additionally, its use could extend to earlier lines of treatment as research continues to determine the optimal sequencing and combination strategies. Due to its mechanism of action, there is potential for elranatamab to be employed not only as a monotherapy but also as part of combination regimens aimed at overcoming resistance mechanisms and deepening therapeutic responses.

Clinical Trials and Studies
Clinical trial evidence from studies such as the Phase 2 MagnetisMM-3 trial has been central to establishing the clinical utility of elranatamab. In these trials, subcutaneous doses following a step-up priming regimen have resulted in objective response rates (ORR) of around 61% in heavily pretreated patients, with many achieving deep responses including complete and very good partial responses. The design of these trials has focused on both efficacy and safety endpoints, monitoring the rate, severity, and duration of adverse events such as CRS and neurotoxicity. Advanced monitoring strategies have been implemented in the trials to adjust dosing schedules and improve patient outcomes.

The ongoing trials are also assessing the transition from once-weekly to biweekly dosing in patients who respond well to therapy. This change in administration frequency has the potential to maintain efficacy while reducing cumulative toxicity and improving long-term tolerability. Moreover, the trials have provided valuable insights into pharmacokinetics and pharmacodynamics, informing on cytokine release levels, time to maximum concentration (Tmax), and half-life parameters, all of which influence dosing strategies and overall therapeutic management. The inclusion of multiple patient populations, such as those with or without prior BCMA-directed therapy, further expands the understanding of elranatamab’s clinical profile and optimizes its therapeutic scope.

Challenges and Future Prospects
While elranatamab represents a significant advancement in multiple myeloma therapy, several challenges remain in its clinical application, and ongoing studies continue to refine its use and maximize patient benefit. Both immediate issues related to safety and tolerability, as well as future considerations implemented through innovative research, are areas of active focus.

Current Challenges in Use
One of the significant challenges in using bispecific antibodies as a therapeutic class, including elranatamab, is mitigating adverse events. Cytokine release syndrome (CRS) is one of the most common adverse events observed in the clinical studies of elranatamab, although the majority of cases are Grade 1 or 2. The initial administration necessitates a step-up dosing strategy to reduce the intensity of immune activation and to manage symptoms effectively. In addition to CRS, other potential toxicities such as neurotoxicity and cytopenias require careful monitoring. The management protocols, including guidelines on dose withholding and supportive care measures, have been detailed in the drug’s administration guidelines to ensure patient safety.

Moreover, the long-term durability of responses remains a topic of clinical interest. Although early data from the MagnetisMM-3 trial have shown promising ORR and durability of response in many patients, continued follow-up is needed to assess long-term overall survival, duration of response, and progression-free survival. The optimal patient selection criteria are also under investigation; understanding the molecular and clinical biomarkers that predict response to elranatamab is critical to maximize its therapeutic benefits while minimizing unnecessary exposure in non-responsive patients.

The immunogenicity of protein-based therapeutics is yet another area of concern. While elranatamab has been engineered to reduce the formation of anti-drug antibodies, ongoing studies need to monitor immunogenic responses over extended periods, as these could impact both efficacy and safety profiles. Regulatory challenges, such as confirming clinical benefit in larger and diverse patient populations and aligning with accelerated approval requirements, also pose hurdles that need to be addressed by further studies and real-world data collection.

Future Research Directions and Potential
Future research on elranatamab is poised to extend its application not only by optimizing monotherapy regimens but also by combining it with other therapeutic modalities. Several research directions are anticipated in the near future:

• Combination Strategies: Future clinical trials will investigate the potential of combining elranatamab with other agents like checkpoint inhibitors, proteasome inhibitors, or other immunomodulatory drugs. These combinations could provide synergistic effects, potentially overcoming resistance mechanisms and further enhancing the anti-myeloma effect. Several ongoing trials, such as those combining talquetamab with teclistamab or with PD-1 inhibitors, set the stage for analogous studies with elranatamab.

• Improved Dosing Regimens: Efforts to move from weekly subcutaneous dosing to biweekly or even less frequent dosing schedules are under exploration to improve patient convenience and reduce healthcare burden while maintaining efficacy. The transitions observed in studies like MagnetisMM-3 describe the improvements in safety profiles when patients switch dosing intervals after achieving sustained responses.

• Biomarker-Driven Patient Selection: More refined strategies for identifying patients most likely to benefit from elranatamab are in development. Future research will focus on molecular profiling and the identification of predictive biomarkers such as BCMA expression levels, immune cell signatures, and genetic alterations that correlate with treatment response. This personalized therapeutic approach may not only enhance the overall efficacy but also reduce undue toxicity in patients unlikely to benefit.

• Enhanced Drug Engineering: Continued advancements in antibody engineering may lead to modifications that increase specificity, reduce off-target effects, or further extend half-life. Structural adjustments and alternative formulations could also enhance tissue penetration and improve overall clinical outcomes. The development of next-generation bispecific antibodies that build upon the successes of elranatamab is an active area of interest in the biopharmaceutical industry.

• Long-Term Safety and Real-World Effectiveness: As elranatamab gains wider clinical use, long-term studies and post-market surveillance will be essential to gather real-world data on its safety and effectiveness. This will inform clinicians about the best practices for its use in various patient demographics, especially in a disease as complex as multiple myeloma where resistance and adverse events are ongoing concerns. Researchers and regulatory agencies are keen on collaborative efforts to continuously update dosing guidelines and explore measures to mitigate adverse events in real-world settings.

• Expanding Indications: While its current primary indication is relapsed or refractory multiple myeloma, there is potential for elranatamab to be evaluated in earlier lines of therapy or even in combination treatments for newly diagnosed patients. The ability to harness an immune-mediated mechanism of action could also see utility in other plasma cell dyscrasias or even in solid tumors, provided that relevant tumor-specific surface antigens can be identified.

Conclusion
In conclusion, elranatamab is classified within the therapeutic class of bispecific antibodies and functions as a T-cell engaging antibody specifically designed to target multiple myeloma cells by linking BCMA on malignant plasma cells with CD3 on T cells. This mechanism not only assigns it to the molecular immunotherapeutic category but also highlights its potential to redirect the innate cytotoxic power of T cells to eliminate tumor cells. The extensive clinical data, primarily provided by studies such as the Phase 2 MagnetisMM-3 trial, support its use in relapsed or refractory multiple myeloma, offering a valuable treatment option for patients who are refractory to conventional therapies.

From a therapeutic standpoint, elranatamab shares similarities with other bispecific T-cell engagers such as teclistamab and blinatumomab, yet its unique target antigen (BCMA) and the specific engineering behind its mode of action provide distinct clinical advantages. It is designed to achieve potent immune activation while employing dosing strategies—such as step-up priming—to manage adverse events like cytokine release syndrome effectively. The clinical implications of elranatamab extend beyond monotherapy; it is under active investigation in combination regimens and may eventually be integrated into earlier lines of treatment based on further research and biomarker-driven patient selection.

Despite its outstanding promise, challenges remain regarding safety management, long-term durability of responses, and immunogenicity. Future research is likely to address these challenges through optimized dosing, advanced combination strategies, enhanced patient stratification, and next-generation antibody engineering, which altogether could broaden its clinical application and further establish its role in the revolution of immunotherapy for multiple myeloma. Overall, elranatamab stands as a highly promising therapeutic agent that exemplifies the modern advances in antibody-based immunotherapy, heralding a new era of targeted, immune-directed treatments in 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.