What are the therapeutic applications for PRAME modulators?

11 March 2025
Introduction to PRAME

Definition and Function of PRAME
PRAME, which stands for “Preferentially Expressed Antigen in Melanoma”, is a cancer/testis antigen originally identified due to its high expression in melanoma cells relative to normal tissues. It belongs to a unique class of proteins that are characteristically silent in most adult tissues but are aberrantly expressed in various cancers. Functionally, PRAME is known to act as a dominant repressor of retinoic acid (RA) signaling – a pathway that is essential for cell differentiation, proliferation arrest, and apoptosis. This repressive action directly contributes to tumor progression by preventing the normal differentiation of cells, fostering conditions in which cancer cells can continue to multiply unchecked. Additionally, PRAME has roles in transcriptional regulation, the maintenance of telomere integrity through protein interactions, and modulation of the immune response, highlighting its multifunctional nature in cancer biology.

Role of PRAME in Cancer and Other Diseases
PRAME is implicated in the pathogenesis of various cancers, including melanoma, acute myeloid leukemia (AML), breast cancer, and several solid tumor types. In cancer cells, its overexpression has been linked to enhanced proliferative capacity, metastatic potential, and resistance to apoptosis. For instance, PRAME’s ability to repress retinoic acid receptor (RAR) target genes not only impedes cellular differentiation but also may contribute to the failure of conventional differentiation therapies in cancers such as acute promyelocytic leukemia (APL). Beyond its oncogenic roles, PRAME is recognized by cytotoxic T lymphocytes (CTLs) when its peptides are presented on major histocompatibility complexes (MHCs) on tumor cells. This particular feature makes it an attractive immunotherapeutic target. Furthermore, despite its primary association with tumors, emerging research suggests that PRAME’s regulation of immune pathways might have implications in modulating inflammatory responses, which could extend its relevance to other pathological conditions beyond cancer.

PRAME Modulators

Types of PRAME Modulators
Several classes of therapeutic agents, collectively referred to as PRAME modulators, have emerged primarily in the context of immunotherapy. These include:

• T-lymphocyte cell therapies: Agents such as Zelenoleucel and MT-401-OTS represent T-lymphocyte cell therapies that are engineered to target multiple tumor-associated antigens, including PRAME. These cell therapies aim to replace or augment a patient’s T-cell repertoire to specifically recognize and kill tumor cells expressing PRAME.

• TCR (T-cell receptor) therapies: A number of PRAME-targeted TCR therapies (for example, those developed by Max-Delbrück-Centrum für Molekulare Medizin (MDC) and TScan Therapeutics, Inc.) have been developed to leverage the high specificity of engineered T cells. These agents are designed to recognize PRAME-derived peptides in the context of specific HLA molecules, thereby initiating an immune response predominantly against tumor cells.

• Bispecific antibodies/T cell engagers: Modulators such as IMA-402 and CDR-813 employ bispecific T-cell engagers (BiTEs) that bind simultaneously to CD3 on T cells and to PRAME on tumor cells. This dual specificity bridges T cells to the tumor microenvironment, enhancing their cytotoxic effect on cells expressing PRAME.

• Diagnostic and prognostic modulators: Beyond direct therapeutic interventions, there are novel diagnostic kits and approaches utilizing PRAME-specific primers and probes, which can be used to detect PRAME gene expression in various cancers. Although these are not direct modulators of function, they play a crucial role in identifying patients who may benefit from PRAME-targeted therapies.

Mechanism of Action
The mechanisms by which PRAME modulators exert their therapeutic effects are multifaceted and depend on the type of modulator used. For cell-based therapies and TCR approaches, the primary mechanism involves the re-direction of the immune system toward cancer cells that aberrantly express PRAME. Once the patient’s T cells are genetically engineered to express PRAME-specific receptors or to be activated by bispecific antibodies, these cells can recognize and bind to cancer cells presenting PRAME-derived peptides on their surface. This recognition leads to the activation of cytotoxic pathways, release of cytokines, and the subsequent killing of the tumor cells.

Furthermore, some therapies operate by “unmasking” the tumor cells to the immune system. By modulating the expression of PRAME or altering its interaction with retinoic acid signaling pathways, these modulators may restore normal RA signaling, thereby promoting differentiation and apoptosis of the malignant cells. In addition, by engaging immune effector functions, these agents can induce immunologic cytotoxicity and amplify T lymphocyte responses, thus overcoming tumor-induced immunosuppressive mechanisms that lead to immune evasion.

The overall biochemical mechanism involves PRAME modulators influencing the downstream signaling cascades that regulate transcription, cell cycle progression, and even the recruitment of ubiquitin ligases that target key proteins for degradation. This multi-step disruption of cancer cell homeostasis results in both direct cell death and an enhanced immune-mediated clearance of tumor cells, thereby amplifying the therapeutic effect.

Therapeutic Applications of PRAME Modulators

Cancer Therapy
Cancer therapy represents the most extensively explored application for PRAME modulators. Given that PRAME is aberrantly overexpressed in various cancers including melanoma, AML, breast, lung, and uveal melanomas, it serves as a prominent tumor-associated antigen – an ideal target for immune-based therapies. The following points detail the multifaceted roles of PRAME modulators in cancer therapy:

• Immunotherapy through adoptive T-cell therapy:
Adoptive cell therapies, such as those using engineered TCR-T cells (e.g., Zelenoleucel and MT-401-OTS), involve the extraction and genetic modification of a patient’s T cells to express receptors specific for PRAME. Once re-infused, these modified T cells home to the tumors expressing PRAME and directly mediate cytotoxicity. Early clinical trials have shown promising responses with objective response rates ranging up to 50–80% in selected patient cohorts, indicating a robust potential for these approaches. Moreover, these therapies often involve strategies to enhance cellular persistence and expansion in vivo, such as the addition of a CD8 co-receptor in next-generation approaches (IMA203CD8).

• Bispecific antibodies and T-cell engagers:
Drugs such as IMA-402 and CDR-813 utilize bispecific antibodies that link T cells (via CD3) to cancer cells expressing PRAME. The simultaneous engagement ensures that T cells are brought in close proximity to tumor cells, facilitating a potent immune response. In addition to triggering T-cell activation, these antibodies help overcome tumor immune evasion by bypassing the need for antigen presentation through classical MHC pathways, which may be downregulated in some malignancies.

• Restoration of RA signaling:
Since PRAME functions as a repressor of retinoic acid signaling, modulators that inhibit or neutralize PRAME can potentially restore RA-mediated cell differentiation and apoptosis. This approach is especially relevant in tumors that have shown resistance to retinoic acid-based therapies. By counteracting the inhibitory effects of PRAME, these agents can sensitize tumors to RA, promoting differentiation and reducing proliferation.

• Combination therapies for enhanced efficacy:
The immunogenic nature of PRAME makes it a promising candidate for combination therapy with immune checkpoint inhibitors, chemotherapy, or even other targeted modalities. For instance, early-phase clinical trials are exploring the use of PRAME modulator-based adoptive T-cell therapies in combination with PD-1 inhibitors to improve durability of response and overcome resistance mechanisms within the tumor microenvironment. These combinations have the potential to amplify the antitumor immune response while reducing the likelihood of tumor escape through adaptive resistance pathways.

• Biomarker-guided therapy and personalized medicine:
PRAME expression levels in tumors can serve as a biomarker to stratify patients who would benefit most from PRAME-targeted interventions. In such situations, diagnostic tools employing PRAME-specific primers and probes are used to detect high PRAME expression, which may predict responsiveness to immunotherapies. The integration of such diagnostic approaches with therapeutic strategies allows for a more personalized treatment regimen, maximizing therapeutic benefits while minimizing unnecessary toxicity.

Autoimmune Diseases
While the primary focus of PRAME modulators to date has been on cancer therapy, there is an emerging interest in the potential modulation of immune responses in autoimmune diseases. Although direct evidence for PRAME modulators in the treatment of autoimmune conditions is currently limited, several aspects provide rationale for exploration in this area:

• Immune regulatory role of PRAME:
PRAME has been implicated in the modulation of immune responses, partly due to its structural similarities with proteins involved in innate immune recognition, such as Toll-like receptors. This structural similarity suggests that PRAME might participate in pathways that regulate inflammation. By modulating PRAME, it is conceivable to adjust the immune system’s activation threshold, thereby potentially ameliorating aberrant immune responses observed in autoimmune conditions.

• Potential for immune recalibration:
In autoimmune diseases, where the immune system erroneously attacks self-tissues, precise modulation to restore tolerance is a key therapeutic goal. PRAME’s role as an immune-modulatory factor could be harnessed to tip the balance in favor of regulatory pathways rather than pro-inflammatory ones. Although experimental data are still emerging, modulation of PRAME may allow for a recalibration of T-cell responses, reducing autoimmunity without broadly suppressing the immune system. Future research may establish protocols where low-dose or targeted PRAME modulators work in synergy with other immunomodulatory agents to achieve this balancing act.

• Combining immunotherapy approaches:
Given that bispecific antibodies and TCR therapies designed for cancer immunotherapy rely on redirecting T-cell responses, similar strategies could potentially be retooled in autoimmune disorders where modulation rather than outright cytotoxicity is desired. For example, engaging regulatory T cells (Tregs) via PRAME-targeted approaches might help control inflammation and auto-reactivity. Although such applications remain exploratory at this stage, the conceptual framework provided by cancer immunotherapy can guide future research into autoimmune applications.

Other Potential Applications
Beyond the realm of cancer and autoimmune diseases, PRAME modulators hold promise for several other applications:

• Diagnostic applications and prognostication:
The detection of PRAME expression in various tissues is not only useful for guiding therapy but also plays a significant role in disease diagnosis and prognosis. Diagnostic kits comprising PRAME-specific primers and probes can be employed to assess tumor load and determine disease progression. Such diagnostic tools may be especially useful in monitoring treatment responses or in early detection of relapse, thereby enhancing overall patient management.

• Combination with conventional therapies:
PRAME modulators can also be combined with conventional chemotherapeutic agents or radiation therapy to enhance overall efficacy. For instance, by restoring RA signaling through PRAME inhibition, tumors that were previously resistant to chemotherapy might become sensitized, thereby increasing the therapeutic index of existing treatments.

• Application in adoptive cell therapy beyond oncology:
The principles underlying PRAME-targeted adoptive cell therapies might be extended to other diseases where precise cell-mediated immunity is desired. Although the bulk of research is focused on oncology, the techniques developed for isolating and engineering T cells to target specific antigens could be leveraged for infectious diseases or conditions involving chronic inflammation, where a tailored immune response is beneficial.

Research and Development

Current Clinical Trials
There is a growing number of clinical trials exploring PRAME modulators, reflective of the increasing interest in targeting tumor-specific antigens for cancer immunotherapy. Several investigational products are in various phases of clinical development:

• Zelenoleucel – A T-lymphocyte cell therapy candidate developed by Marker Therapeutics, Inc., which is currently being assessed in Phase 2 trials for its ability to target multiple antigens including PRAME.

• MDG-1011 – Developed by Max-Delbrück-Centrum für Molekulare Medizin (MDC), this agent employs TCR therapy specifically targeting PRAME and is in Phase 2 of clinical development for hematologic diseases.

• MT-401-OTS – A T-lymphocyte cell therapy designed to target a combination of antigens including PRAME, currently in Phase 1 clinical trials, indicating the early translational potential of these complex immunotherapeutic approaches.

• IMA-402 and CDR-813 – These bispecific modalities, acting as T-cell engagers by binding both CD3 and PRAME, are in Phase 1/2 or preclinical stages. These candidates represent a novel class of medicines that harness the body’s immune system to achieve targeted cytotoxicity against PRAME-expressing tumors.

• TSC-203-A0201 – A PRAME-targeted TCR therapy in Phase 1 demonstrating early promise for direct engagement of T cells in tumor elimination.

The diversity of modalities in current clinical trials underscores the strategic importance of PRAME as a therapeutic target and reflects the multi-angle approach adopted by various research groups to overcome tumor immune evasion. Moreover, these trials are designed with built-in biomarker assessments to confirm PRAME expression in patient tumors and to correlate clinical responses to modulator activity, thereby establishing robust links between target expression and therapeutic efficacy.

Challenges in Development
Despite significant progress, several challenges remain in the development of PRAME modulators:

• Antigen heterogeneity and expression variability:
One of the major obstacles is the variability in PRAME expression among different tumor types and even within subpopulations of the same tumor. This heterogeneity poses challenges for both the efficacy and the safety of PRAME-targeted therapies since adequate expression is required for optimal therapeutic response, and off-target effects may arise in tumors with low or inconsistent PRAME expression.

• Manufacturing and scalability of cell therapies:
For adoptive cell therapies and engineered T-cell products, issues related to large-scale manufacturing, quality control, and reproducibility are critical. Variability in cell product formulations and challenges in maintaining consistent expression of engineered receptors may impact clinical outcomes. Regulatory hurdles and high production costs also add layers of complexity to these advanced therapies.

• Immunogenicity and resistance:
The interplay between tumor cells and the immune system is highly dynamic. While PRAME modulators have shown promise, there is always a risk that tumors may develop resistance mechanisms – such as antigen loss, altered antigen processing, or adaptive immune resistance – to evade targeted therapies. In addition, repeated stimulation of T cells may lead to exhaustion or increased immunogenicity that could counteract therapeutic benefits.

• Safety and off-tumor toxicity:
Although PRAME is largely restricted to cancer cells, there remains a need for thorough evaluation to ensure that targeting PRAME does not result in adverse effects in normal tissues, particularly given its potential low-level expression in certain normal cells or during embryonic development. Strategies to mitigate on-target off-tumor toxicity, such as dose optimization and improved cell engineering techniques, are under active investigation.

Future Directions and Implications

Emerging Research
Emerging research continues to refine our understanding of PRAME and its role in tumor immunobiology. Notable areas of future exploration include:

• Combination therapies:
There is an increasing emphasis on using PRAME modulators in conjunction with other immunotherapies such as checkpoint inhibitors, cytokine therapies, or additional targeted agents. The synergistic potential of such combination regimens may lead to enhanced tumor eradication, overcoming resistance seen with monotherapies.

• Advanced cell engineering techniques:
Next-generation approaches are focusing on enhancing the persistence and functionality of PRAME-targeted T cells. Innovations such as dual receptor expression (e.g., co-transduction with CD8 receptors in IMA203CD8) and optimized cell manufacturing protocols aim to improve both efficacy and safety profiles.

• Personalized medicine and biomarker-driven strategies:
With diagnostic tools based on PRAME expression advancing rapidly, the integration of personalized medicine frameworks is anticipated. Such strategies involve using PRAME expression levels to stratify patients and tailor therapies according to individual tumor antigen profiles. The utilization of high-throughput sequencing, spatial transcriptomics, and single-cell analysis further supports these efforts.

• Exploration in non-oncologic indications:
Although early investigations focus on oncology, the immunomodulatory roles of PRAME could eventually provide insights into its utility in autoimmune diseases and other conditions characterized by dysregulated immune responses. With a deeper understanding of PRAME’s signaling networks, targeted modulation could one day extend beyond cancer to treat disorders with an inflammatory or autoimmune component.

Potential Impact on Healthcare
The clinical translation of PRAME modulators stands poised to revolutionize targeted cancer immunotherapy. Their potential impact encompasses:

• Improved overall survival and quality of life in cancer patients:
By specifically targeting tumor-associated antigens such as PRAME, these modulators offer the promise of highly effective and less toxic therapies compared to conventional chemotherapy. Early-phase clinical trials demonstrating high objective response rates further validate the potential for improved clinical outcomes.

• Reduction of off-target toxicities:
Targeted immunotherapy inherently minimizes systemic side effects by focusing the immune response on PRAME-expressing cells. Such specificity is expected to reduce the adverse event profile and improve the tolerability of therapies, thereby enhancing overall patient quality of life.

• Advancement in personalized medicine:
The development of diagnostic assays to assess PRAME expression paves the way for personalized treatment regimens. This biomarker-guided approach ensures that patients receive therapies most likely to benefit them and facilitates monitoring of treatment response and disease progression.

• Broadening treatment horizons beyond oncology:
Even though the majority of current research is oriented toward cancer, the insights gained through PRAME modulation may eventually lead to novel interventions in autoimmune diseases and other immune-related disorders. The translational potential of such findings could ultimately broaden therapeutic options for a range of pathologies.

• Economic and regulatory considerations:
As manufacturing processes become streamlined and clinical data mature, PRAME-targeted therapies have the potential to become cost-effective additions to the therapeutic arsenal. Regulatory agencies are increasingly supportive of innovative immunotherapies, which might accelerate the approval process and bring these treatments to market faster.

Conclusion
In summary, PRAME modulators represent a highly promising frontier in targeted therapeutics, particularly within oncology. PRAME—as a cancer/testis antigen with critical roles in tumor biology such as the repression of retinoic acid signaling—provides an almost ideal target for immune-mediated therapies. The diversity of modulators under development, ranging from T-cell receptor (TCR) therapies and adoptive T-cell therapies like Zelenoleucel and MT-401-OTS, to bispecific T-cell engagers such as IMA-402 and CDR-813, underscores the multi-pronged approach currently being pursued.

Therapeutically, these agents offer a multifaceted mechanism of action: they not only directly target tumor cells through highly specific immune recognition but also restore critical cellular pathways—such as RA signaling—that induce differentiation and apoptosis. Their use in cancer therapy has demonstrated significant promise in early-phase clinical trials, with encouraging objective response rates and favorable safety profiles indicating a potential shift away from traditional cytotoxic chemotherapies.

While the primary applications of PRAME modulators lie in oncology, there exists an intriguing possibility that modulation of PRAME could extend to autoimmune diseases by recalibrating aberrant immune responses, although this area remains largely exploratory at present. Diagnostic applications related to PRAME detection further emphasize its role in personalized medicine, allowing for the precise stratification of patient populations that may benefit most from such targeted interventions.

Despite these promising advances, the field faces challenges, including variability in antigen expression, risks of immunogenicity and resistance, and the complexities of manufacturing cell-based therapies. Overcoming these hurdles will involve innovations in cell engineering, combination therapy strategies, and improved manufacturing protocols. Emerging research that leverages advanced genomic and proteomic approaches holds the potential to refine these therapies further and expand their application to a broader range of diseases.

In conclusion, PRAME modulators provide a general framework for rethinking cancer therapy from a targeted, immune-based perspective. Specifically, they enable the restoration of normal cellular pathways, increase tumor immunogenicity, and offer substantial improvements in patient outcomes by harnessing the precision of the immune system. With ongoing clinical trials and substantial research endeavors driving the field forward, PRAME modulators are likely to have a transformational impact on healthcare, leading not only to more effective cancer treatments but also paving the way for future therapeutic applications in autoimmune conditions and beyond. The continued advancement in this area holds promise for ushering in a new era of personalized, targeted medicine that maximizes therapeutic efficacy while minimizing systemic toxicity.

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.