What is the therapeutic class of Entinostat?

7 March 2025
Introduction to Entinostat
Entinostat is an orally available small‐molecule drug candidate that has captured considerable attention due to its unique ability to modulate the epigenetic landscape required for both tumor cell regulation and immune system engagement. Over more than two decades of research, entinostat has emerged as one of the leading agents in the field of epigenetic therapy for solid tumors and hematologic malignancies. By targeting key enzymes involved in chromatin organization, entinostat can reverse aberrant gene silencing—a hallmark of many cancers—and thereby restore normal gene transcription patterns. In addition to exerting direct antitumor effects, entinostat has gained popularity for its potential to alter the tumor microenvironment (TME), including modulation of immune regulatory cells, and is often employed—either alone or in combination with other agents—as part of novel cancer immunotherapeutic strategies.

Chemical Composition and Structure
Chemically, entinostat is a synthetic benzamide derivative, designed to serve as a highly specific inhibitor of class I histone deacetylases (HDACs) such as HDAC1, HDAC2, and HDAC3. This molecular configuration distinguishes entinostat from other inhibitors that possess broad spectrum inhibitory activity toward multiple HDAC isoforms. The benzamide scaffold is central to its selective binding characteristics; subtle modifications to its aromatic and side-chain features were engineered to optimize potency and pharmacokinetic properties. The compound’s design ensures that its inhibitory effect is largely confined to the HDAC isoforms most crucial to driving the aberrant epigenetic modifications in aggressive tumors, thereby promoting a favorable safety profile and minimizing off-target toxicities. In many synapse‐sourced publications, entinostat’s structure is cited as the key element underlying its pharmacological profile, where its long half‐life enables continuous exposure to pharmacologically active concentrations in patients.

Development and Approval History
Over the past several years, research on entinostat has traversed a long pathway from preclinical validation to advanced clinical trials. Early preclinical studies provided strong evidence of its efficacy in reversing gene repression and inhibiting tumor growth, while also highlighting its immunomodulatory potential. Subsequent phase I and phase II trials established that entinostat has a favorable safety profile in over 800 to 900 cancer patients, with promising antitumor activity demonstrated in diseases such as breast cancer, lung cancer, and certain hematologic malignancies.
The development history of entinostat is strongly tied to efforts by companies such as Syndax Pharmaceuticals, which positioned entinostat as the lead product candidate among a portfolio of epigenetic modulators. It has gained breakthrough therapy designation by the U.S. Food and Drug Administration (FDA) for advanced hormone receptor–positive (HR+) breast cancers—a designation underscoring its potential role as a treatment option in this patient population. In various clinical programs, including the pivotal E2112 trial and other combination strategies (e.g., with exemestane or immune checkpoint inhibitors), entinostat is studied intensively to address unmet clinical needs in both endocrine-resistant and immuno-responsive tumors. Research continues to extend its application to other malignancies and to optimize combination regimens—indicative of a rich, evolving history in the oncology research arena.

Pharmacological Classification
Pharmacologically, entinostat falls into the therapeutic class of selective epigenetic modulators, specifically as a class I histone deacetylase (HDAC) inhibitor. To understand its classification fully, it is essential to consider both the general definition of its therapeutic category and the specific characteristics that delineate entinostat within that group.

Definition of Therapeutic Class
The term “therapeutic class” refers to a group of drugs that share similar mechanisms of action, pharmacological effects, and clinical applications. In the context of cancer therapeutics, many agents are classified based on their target enzymes or pathways. Epigenetic modulators, for example, are a diverse set of drugs that alter gene expression without directly modifying the DNA sequence. Among these, histone deacetylase inhibitors (HDACi) act by interfering with the function of histone deacetylases—enzymes that normally remove acetyl groups from histone proteins, leading to tighter chromatin packing and reduced gene expression. By inhibiting HDAC activity, these drugs maintain a more open chromatin structure that permits the transcription of genes that may have been silenced during tumorigenesis. This mechanism provides an effective means to restore the expression of tumor suppressor genes and modulate cellular differentiation or apoptosis, placing HDAC inhibitors squarely in the broader category of epigenetic therapeutics.

Entinostat's Therapeutic Class
Entinostat is categorically defined as a class I selective HDAC inhibitor because it primarily suppresses the activity of HDAC1, HDAC2, and HDAC3 enzymes. This selective inhibition is particularly important, as these isoforms are frequently implicated in the aberrant epigenetic repression observed in a variety of malignancies. By targeting class I HDACs, entinostat can suppress oncogenic transcriptional programs that contribute to tumor growth, metastasis, and immune evasion.
The therapeutic class of HDAC inhibitors encompasses both pan-HDAC inhibitors, which affect multiple classes of HDAC enzymes, and selective agents like entinostat that narrow their activity to specific isoforms. Entinostat’s strategic selectivity contributes not only to its antitumor potency but also to its enhanced safety profile, as it tends to spare cytotoxic T cells from adverse cytotoxic effects—a benefit that is particularly desirable when used in immunotherapeutic combinations. Furthermore, studies published on synapse have elucidated that, by modulating HDAC activity, entinostat can influence cellular processes such as the acetylation state of transcription factors (e.g., STAT3) and key regulators like FoxP3, further validating its classification as an agent that can reprogram the epigenetic status of both cancer and immune cells. This unique combination of selective epigenetic modulation and immunomodulatory function has propelled entinostat to the forefront of novel oncology treatments.

Mechanism of Action
Understanding the mechanism of action of entinostat involves a multi‐layered exploration—from its molecular targets and pathways to the resulting biological effects that culminate in its antitumor efficacy.

Molecular Targets
At a fundamental level, entinostat’s direct molecular targets are class I HDAC enzymes, particularly HDAC1, HDAC2, and HDAC3. These enzymes are crucial in the removal of acetyl groups from lysine residues on histone proteins, which, in turn, influences chromatin structure and gene expression. In tumors, the overactivity of these HDACs often leads to transcriptional repression of tumor suppressor genes, facilitating unchecked proliferation and evasion of programmed cell death.
By binding to the catalytic pocket of class I HDACs, entinostat prevents the deacetylation process, which results in increased acetylation levels of histone proteins such as H3 and H4. This hyperacetylation effect relaxes the chromatin structure, thereby allowing a reactivation of previously silenced genes. Importantly, entinostat has been demonstrated in multiple studies to specifically reduce the expression of FoxP3 in regulatory T cells (Tregs) through direct transcriptional inhibition, a mechanism that involves partial modulation of STAT3 acetylation. This biochemical cascade underlies entinostat’s ability to disrupt the immune-suppressive functions of Tregs while sparing, or even enhancing, the activity of effector T cells.
Beyond histones, entinostat can also exert effects on nonhistone substrates. For example, its influence on transcription factors and cell cycle regulators has been linked to enhanced susceptibility of tumor cells to immune cell recognition and apoptotic signals. These molecular interactions are pivotal in turning the intrinsic properties of malignant cells against them, setting the stage for both direct cytotoxic effects and an improved immunologic response to the tumor.

Biological Effects
The inhibition of class I HDACs by entinostat triggers a cascade of molecular events that collectively contribute to its antitumor activity. The primary biological effect is the reactivation of key genes that play central roles in cell cycle regulation, differentiation, and apoptosis. With chromatin in a more open configuration, genes that encode tumor suppressors are re-expressed, leading to a cessation of uncontrolled proliferation and, ultimately, the induction of cell death pathways in tumor cells.
Furthermore, entinostat’s role as an immunomodulatory agent has been particularly highlighted in preclinical models. Through the downregulation of FoxP3 in Tregs, entinostat reduces immune suppression within the TME, thus permitting an enhanced infiltration and activation of CD8+ cytotoxic T cells. This immunogenic remodeling of the TME results in improved antigen presentation and increased expression of major histocompatibility complex (MHC) proteins, making cancer cells more "visible" to the immune system. In breast cancer models, as well as in other solid tumor types, evidence shows that the use of entinostat leads to increased sensitivity to T cell–mediated lysis and even potentiates the action of immune checkpoint inhibitors.
On a broader scale, entinostat also displays synergy with various chemotherapeutic agents. For example, its use in combination with doxorubicin, aromatase inhibitors, or even targeted immunotherapies has demonstrated enhanced overall antitumor efficacy. This synergistic potential is partially attributable to the capacity of entinostat to induce long-lasting changes in gene expression patterns and to alter cellular signaling pathways that are critical for tumor survival. The net biological effect is a multi-pronged attack on the tumor: direct inhibition of cell growth, induction of apoptosis, reversal of immune tolerance, and sensitization of tumor cells to additional therapeutic modalities.

Clinical Applications
The translational significance of entinostat lies in its potential to transform clinical treatment strategies through both its direct cytotoxic effects on tumor cells and its pivotal role in immune modulation.

Approved Uses
Although entinostat is still under clinical evaluation and has not yet received full regulatory approval as a monotherapy, it has shown significant promise as part of combination regimens. The U.S. FDA’s breakthrough therapy designation granted in the context of advanced HR+ breast cancer underscores the clinical potential of entinostat in this setting.
In numerous phase II clinical trials, the combination of entinostat with endocrine therapies—most notably exemestane—has demonstrated a clinically meaningful overall survival (OS) benefit in patients with advanced breast cancer resistant to first-line endocrine treatments. The promising results from these trials have led to an ongoing phase III registration trial (E2112) that aims to secure a potential new treatment option for patients with hormone receptor–positive, HER2-negative breast cancer. Moreover, data emerging from clinical studies have highlighted its favorable safety profile compared with other HDAC inhibitors, further supporting its candidacy in challenging clinical scenarios where toxicity is a major concern.

Ongoing Clinical Trials
A multitude of clinical investigations are ongoing to evaluate the efficacy and safety of entinostat across various cancer types. One key focus has been on its combination with endocrine therapies in HR+ breast cancer, a strategy that seeks to reverse endocrine resistance via epigenetic reprogramming. In addition, synergistic combinations with immunotherapies—such as checkpoint inhibitors (e.g., pembrolizumab and atezolizumab)—are currently being evaluated in both non–small cell lung cancer (NSCLC) and melanoma, among other immuno‐responsive tumors.
There is also interest in exploring the application of entinostat in pediatric tumors; a phase I study in children and adolescents with recurrent or refractory solid tumors, including CNS tumors, established its tolerability in the pediatric population and encouraged further development. Another promising area of investigation is the use of entinostat in combination with cancer vaccines and cytokine therapies (e.g., IL-15 superagonists) to potentiate antitumor immune responses by reducing regulatory T cell and myeloid-derived suppressor cell (MDSC) activity in the TME. Clinical studies designed to evaluate entinostat’s role in synergistic regimens underscore its versatility as both an epigenetic modulator and an immunotherapeutic enhancer.

Challenges and Future Directions
Despite the advanced stage of clinical evaluation and encouraging preliminary data, several challenges remain for entinostat, as is common with many agents operating in the relatively new field of epigenetic therapy. Understanding these challenges and defining avenues for future research are essential for the continued development and eventual integration of entinostat into routine clinical practice.

Current Challenges
One of the central challenges with entinostat, and HDAC inhibitors in general, is the sometimes unpredictable correlation between dose and clinical efficacy. Early phase trials have indicated that entinostat’s efficacy is not strictly dose-dependent, suggesting that conventional dosing strategies based solely on maximum tolerated dose (MTD) may not be optimal. Instead, identification of robust biomarkers that can accurately predict patient response and guide dose optimization is urgently needed.
Another challenge pertains to the complex pharmacodynamic effects of epigenetic modulation. While increased histone acetylation generally correlates with the reactivation of tumor suppressor genes, there is significant heterogeneity in gene expression responses across different tumor types and patients. This variability complicates the task of defining dosing schedules that achieve consistent therapeutic effects without incurring undue side effects. Although entinostat’s safety profile has been favorable in many trials, the long half-life of the drug—which is beneficial for continuous target inhibition—also introduces challenges in terms of potential cumulative toxicity and managing adverse events, particularly in combination regimens.
Moreover, while entinostat shows promising immunomodulatory effects, the translation of these effects into consistent clinical efficacy remains to be definitively established. The interplay between entinostat-induced epigenetic changes and the immune system is complex, involving a fine balance between enhancing antitumor immunity and avoiding inadvertent suppression of beneficial immune responses. Dissecting these mechanisms and integrating them with other immunotherapeutic strategies represents an ongoing challenge.
Lastly, because entinostat is still in clinical development, regulatory hurdles, as well as issues related to intellectual property, production scalability, and cost-effectiveness, must be addressed to facilitate its transition from clinical trials to widespread clinical use.

Future Research Directions
Looking ahead, future research on entinostat is poised to focus on several interrelated objectives. First and foremost, the identification and validation of predictive biomarkers are essential. As entinostat’s efficacy may not correlate linearly with dose, establishing biomarkers that reflect intratumoral histone acetylation levels, immune cell infiltration, and gene expression changes will be critical in refining dosing strategies and patient selection criteria.
Research efforts will also continue to elucidate the detailed molecular mechanisms underlying entinostat’s ability to modulate the TME. Beyond its direct effects on tumor cells, entinostat’s impact on immune subsets—particularly its capacity to inhibit regulatory T cells and MDSCs while simultaneously enhancing cytotoxic T cell function—is a promising area of investigation. Future preclinical studies may delve deeper into these mechanisms to identify combinatorial approaches that can further amplify the immunogenic effects of entinostat.
Moreover, expanding the research on combination therapies is a key future direction. Given the encouraging preclinical and clinical results observed when entinostat is paired with endocrine therapies, immunotherapies, and even chemotherapeutic agents, there is significant rationale to explore additional synergistic combinations. For instance, pairing entinostat with other epigenetic modifiers, radiotherapy, or novel immunomodulators may yield additive—or even synergistic—benefits that radically improve clinical outcomes in treatment‐resistant cancers.
Another promising direction is the exploration of entinostat in the context of personalized medicine. As genomic and proteomic profiling technologies become more advanced and accessible, tailoring entinostat-based therapies to the unique molecular characteristics of each patient’s tumor becomes increasingly feasible. Such an approach could optimize patient outcomes by ensuring that entinostat’s epigenetic modulation is most effectively leveraged in the appropriate biological context.
Furthermore, although current clinical investigations have primarily focused on breast, lung, and select pediatric cancers, future clinical trials may explore the use of entinostat in other malignancies, such as renal cell carcinoma, prostate cancer, and even certain hematologic disorders. Extending the investigation to a broader spectrum of tumor types may reveal additional applications for entinostat that have yet to be fully realized.
On the manufacturing and regulatory fronts, efforts to streamline production and overcome logistical hurdles will be crucial. As entinostat’s development advances into later‐phase trials and potentially enters the market, ensuring cost-effective scalability and addressing any regulatory challenges will be imperative for its successful adoption in clinical practice.

Conclusion
In summary, entinostat is classified therapeutically as a selective class I histone deacetylase inhibitor—an epigenetic modulator endowed with multifaceted antitumor and immunomodulatory properties. Its synthetic benzamide derivative structure underpins its selectivity towards HDAC1, HDAC2, and HDAC3, facilitating hyperacetylation of histones and reversal of aberrant gene silencing that is frequently observed in aggressive tumors. Through its unique molecular actions, entinostat not only reactivates tumor suppressor genes but also reconditions the tumor microenvironment by diminishing the immunosuppressive influence of regulatory T cells and myeloid-derived suppressor cells. This dual mechanism has positioned it as a key agent in combination therapies, particularly in the setting of advanced HR+ breast cancer—where it has garnered breakthrough therapy designation by the FDA—and in combination immunotherapy regimens in lung cancer, melanoma, and beyond.

From a clinical perspective, while entinostat is not yet fully approved as a standalone treatment, its advanced-stage clinical trials, especially in combination with endocrine and immunotherapeutic agents, underscore its transformative potential. Ongoing investigations in both adult and pediatric populations further highlight its versatility and promising safety profile. Nevertheless, challenges remain in optimizing dosing regimens, identifying robust predictive biomarkers, and integrating entinostat effectively within multimodal treatment strategies. Future research is focused on overcoming these challenges through comprehensive biomarker identification, mechanistic studies to fine-tune immune and epigenetic interactions, and expansion into innovative drug combinations tailored to personalized medicine approaches.

Overall, entinostat exemplifies the next generation of therapeutic agents in oncology that operate at the intersection of epigenetic modulation and immuno-oncology. Its development history, neatly situated within emerging paradigms of targeted cancer therapy, reflects a compelling journey from concept to clinical application. By continuing to unravel its intricate molecular mechanisms and overcoming current clinical challenges, entinostat holds significant promise to reshape treatment landscapes and improve outcomes for patients across a broad spectrum of cancers. The future of entinostat is bright, with ongoing clinical trials and ongoing research efforts aimed at harnessing its full therapeutic potential while addressing critical challenges in efficacy, safety, and patient-specific responsiveness.

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成