MedPath

Mocetinostat Advanced Drug Monograph

Published:Sep 9, 2025

Generic Name

Mocetinostat

Drug Type

Small Molecule

Chemical Formula

C23H20N6O

CAS Number

726169-73-9

Mocetinostat (MGCD0103): A Comprehensive Monograph on a Spectrum-Selective HDAC Inhibitor in Oncology

Section 1: Introduction and Overview

Introduction to Epigenetic Therapy in Oncology

The field of oncology has progressively evolved beyond cytotoxic and targeted therapies to embrace strategies that manipulate the fundamental regulatory machinery of the cancer cell. Among the most promising of these are epigenetic therapies, which target the mechanisms that control gene expression without altering the underlying DNA sequence.[1] A central process in this regulatory network is the dynamic acetylation and deacetylation of histone proteins. Histone acetyltransferases (HATs) add acetyl groups to lysine residues on histone tails, creating a relaxed chromatin structure that permits gene transcription. Conversely, histone deacetylases (HDACs) remove these acetyl groups, leading to chromatin condensation and transcriptional repression.[2]

In carcinogenesis, the dysregulation of HDACs is a common event. The overexpression or aberrant activity of these enzymes can lead to the inappropriate silencing of critical tumor suppressor genes, which are the body's natural defense against cancer.[5] This epigenetic silencing contributes to uncontrolled cell proliferation, survival, and differentiation, making HDACs a validated and compelling therapeutic target. The development of HDAC inhibitors (HDACis) represents a rational approach to reverse this pathological gene silencing, restore normal cellular function, and thereby inhibit tumor growth.[1]

Positioning Mocetinostat within the HDAC Inhibitor Landscape

Mocetinostat, also known by its developmental code MGCD0103, is an investigational, orally bioavailable, small molecule drug belonging to the 2-aminobenzamide chemical class of HDAC inhibitors.[8] It was rationally designed to offer a more targeted approach compared to the first generation of HDACis. A critical distinction in this therapeutic class is between pan-HDAC inhibitors, such as the FDA-approved agents vorinostat and romidepsin, which inhibit a broad range of HDAC enzymes, and isotype-selective inhibitors.[4] Mocetinostat falls into the latter category, exhibiting potent inhibitory activity against a specific subset of HDACs—namely, the Class I enzymes (HDAC1, HDAC2, HDAC3) and the sole Class IV enzyme (HDAC11).[10] This selectivity was hypothesized to provide an improved therapeutic window by focusing on the HDACs most critically involved in oncogenesis while potentially sparing off-target effects associated with the inhibition of other HDAC classes.

Summary of Developmental Trajectory

The clinical development of Mocetinostat has been extensive and multifaceted, reflecting its broad biological activity. It has been investigated as a monotherapy and in combination regimens across a wide spectrum of malignancies, including both hematologic cancers and solid tumors. Clinical trials have explored its utility in relapsed/refractory lymphoma, myelodysplastic syndromes (MDS), acute myeloid leukemia (AML), metastatic urothelial carcinoma, non-small cell lung cancer (NSCLC), and various sarcomas.[5] This broad investigational scope has generated a wealth of clinical data, painting a complex picture of a drug with undeniable biological potency. However, its journey also illuminates a central challenge in modern drug development: translating potent biological activity into a clinically viable therapeutic with an acceptable risk-benefit profile. The story of Mocetinostat is therefore one of significant promise, marked by compelling efficacy signals in certain contexts, but equally defined by the considerable peril of a challenging toxicity profile that has ultimately limited its clinical translation.

Section 2: Physicochemical Profile and Molecular Identification

A precise and unambiguous identification of a drug molecule is foundational to all research and clinical development. The following table consolidates the key physicochemical and identifying characteristics of Mocetinostat, compiled from authoritative chemical and pharmacological databases. This standardized reference serves to prevent ambiguity and provide a comprehensive chemical datasheet for the compound.

PropertyValueSource(s)
Generic NameMocetinostat5
Synonyms/Code NamesMGCD-0103, MGCD0103, MG01035
DrugBank IDDB118305
CAS Number726169-73-98
UNIIA6GWB8T96J8
Chemical FormulaC23​H20​N6​O5
Molecular WeightAverage: 396.44-396.454 g/mol5
Monoisotopic: 396.169859288 g/mol5
IUPAC NameN-(2-aminophenyl)-4-[[(4-pyridin-3-ylpyrimidin-2-yl)amino]methyl]benzamide8
SMILESC1=CC=C(C(=C1)N)NC(=O)C2=CC=C(C=C2)CNC3=NC=CC(=N3)C4=CN=CC=C48
InChIKeyHRNLUBSXIHFDHP-UHFFFAOYSA-N8
Physical AppearanceWhite to off-white solid powder10
SolubilitySoluble in DMSO and DMF; 0.5 mg/ml in DMF:PBS (pH 7.2) (1:1)10
Storage ConditionsDry, dark, at 0-4 °C (short term) or -20 °C (long term)10

Section 3: Molecular Pharmacology and Mechanism of Action

The pharmacological activity of Mocetinostat is complex, extending beyond its primary epigenetic function to encompass profound effects on the host immune system. Its mechanism of action can be understood as a multi-pronged attack on cancer, involving direct, cell-intrinsic cytotoxicity and indirect, host-mediated immunomodulation.

Selective Inhibition of Class I and IV Histone Deacetylases

Mocetinostat is a potent and isotype-selective inhibitor of the zinc-dependent classical HDACs, specifically targeting Class I (HDAC1, HDAC2, HDAC3) and Class IV (HDAC11) enzymes.[10] It demonstrates negligible activity against Class II HDACs (HDAC4, 5, 6, 7, and 8) at therapeutic concentrations.[13] Cell-free assays have quantified its inhibitory potency (

IC50​ values) against its primary targets as follows:

  • HDAC1: 0.15 µM
  • HDAC2: 0.29 µM
  • HDAC11: 0.59 µM
  • HDAC3: 1.66 µM (Data compiled from 10)

The significance of this target selectivity lies in the biological roles of these specific HDACs in cancer. Class I HDACs are ubiquitously expressed, primarily localized to the nucleus, and are fundamental regulators of cell proliferation and survival.[17] Their overexpression has been directly linked to tumorigenesis and poorer clinical outcomes. For instance, in urothelial bladder cancer, high expression levels of HDAC1 and HDAC2 are associated with higher tumor grades, while HDAC1, HDAC2, and HDAC3 are all found to be overexpressed in tumors compared to normal tissue.[18] By selectively targeting these key oncogenic drivers, Mocetinostat was designed to maximize anti-tumor efficacy while minimizing the off-target effects that might arise from inhibiting Class II HDACs, which have more diverse roles in cellular differentiation and function. The role of HDAC11 is less defined but involves the regulation of hematopoiesis and immune responses, such as controlling IL-10 expression, making its inhibition relevant to Mocetinostat's immunomodulatory profile.[19]

Epigenetic Reprogramming and Cellular Consequences

The core mechanism of Mocetinostat is the direct inhibition of its target HDAC enzymes. By binding to these enzymes, it prevents the removal of acetyl groups from the lysine residues of histone proteins. This leads to an accumulation of acetylated histones (hyperacetylation), particularly on H3 lysine 9 (H3K9) and H3 lysine 27 (H3K27), as demonstrated in breast cancer cell lines.[10] Histone acetylation neutralizes the positive charge of lysine, weakening the electrostatic interaction between the histones and the negatively charged DNA backbone. This results in a more open, relaxed chromatin structure (euchromatin), which allows transcription factors to access gene promoter regions and initiate transcription.[1]

This fundamental epigenetic reprogramming triggers a cascade of downstream anti-cancer effects:

  • Reactivation of Tumor Suppressor Genes: The primary therapeutic hypothesis for Mocetinostat is that it can reactivate tumor suppressor genes that have been epigenetically silenced by the cancer.[5] This includes the upregulation of critical cell cycle inhibitors like p21, which serves as a brake on cell proliferation.[1]
  • Induction of Apoptosis: Mocetinostat robustly induces programmed cell death (apoptosis) in cancer cells. This occurs through caspase-dependent pathways, evidenced by the increased presence of cleaved caspase-3, a key executioner of apoptosis.[8]
  • Cell Cycle Arrest: The drug effectively halts the cancer cell cycle, preventing cells from dividing. Flow cytometry analyses have shown that treatment with Mocetinostat leads to a substantial accumulation of cells in the G1 and G2/M phases of the cell cycle.[8]
  • Induction of Autophagy: In addition to apoptosis, Mocetinostat can trigger autophagy, a process of cellular self-digestion that can also lead to cell death in a cancerous context.[1]

Immunomodulatory Effects

Beyond its direct effects on tumor cells, a growing body of evidence reveals that Mocetinostat has profound immunomodulatory properties that reshape the tumor microenvironment (TME) from an immunosuppressive to an immune-active state. This dual mechanism of action is central to understanding its potential, particularly in combination therapies.

  • Enhancing Tumor Antigen Presentation: Mocetinostat makes tumor cells more "visible" to the immune system. It achieves this by upregulating the expression of genes involved in the antigen presentation machinery, including both Class I and Class II Human Leukocyte Antigen (HLA) family members. It also increases the expression of the Class II transactivator (CIITA), a master regulator of Class II HLA gene expression, and does so synergistically with interferon-gamma (IFN-γ).[21] Enhanced antigen presentation allows cytotoxic T-cells to more effectively recognize and target cancer cells for destruction.
  • Modulation of Immune Checkpoints: Mocetinostat directly upregulates the expression of the immune checkpoint ligand Programmed Death-Ligand 1 (PD-L1) on the surface of tumor cells.[11] While upregulation of an immune-suppressing molecule may seem counterintuitive for an anti-cancer drug, this effect is critically important in the context of combination therapy. The efficacy of anti-PD-1/PD-L1 checkpoint inhibitors is often correlated with the level of PD-L1 expression in the tumor. By increasing PD-L1 levels, Mocetinostat may convert immunologically "cold" tumors (low PD-L1, non-inflamed) into "hot" tumors, effectively "painting a target" on them for partner immunotherapy agents to attack. This provides a powerful rationale for the clinical trials combining Mocetinostat with drugs like durvalumab and nivolumab.
  • Reshaping the Tumor Microenvironment: Mocetinostat actively remodels the cellular composition of the TME. Preclinical and clinical correlative studies have shown that it can:
  • Decrease the populations of immunosuppressive cells, including T-regulatory cells (Tregs) and myeloid-derived suppressor cells (MDSCs), which normally dampen anti-tumor immune responses.[11]
  • Increase the infiltration of tumors by beneficial cytotoxic CD8+ T-cells, the primary effectors of anti-tumor immunity.[11]

This comprehensive immunomodulatory activity explains why Mocetinostat's development has increasingly focused on combinations with checkpoint inhibitors. It does not simply rely on its direct cytotoxic effects; it also acts as an immune primer, creating a TME that is more permissive to a robust and effective anti-tumor immune response.[21]

Non-Oncological Mechanisms of Action

Preclinical research has also uncovered potential therapeutic applications for Mocetinostat outside of oncology, suggesting its mechanisms have broader biological relevance.

  • Cardiorenal Protection: In a rat model of transverse aortic constriction (TAC)-induced pressure overload, Mocetinostat treatment was shown to significantly reduce systolic blood pressure and mitigate adverse cardiac remodeling, including regulating the thickness of the interventricular septum and left ventricular posterior wall.[25] These findings suggest a potential protective effect in hypertension-related cardiac disease.
  • Potential in Osteoarthritis (OA): A high-throughput screen identified Mocetinostat as a potential disease-modifying osteoarthritis drug (DMOAD). In human chondrocytes and other joint tissue cells, it upregulated cartilage signature genes (e.g., KLF4, COL2A1, SOX9) while downregulating genes associated with hypertrophy, inflammation, and catabolism. In a mouse model of OA, administration of Mocetinostat reduced the severity of joint damage and improved pain behaviors.[26]

Section 4: Preclinical Evaluation

The clinical development of Mocetinostat was underpinned by a robust body of preclinical evidence demonstrating its anti-cancer activity and establishing a clear biological rationale for its mechanism of action.

In Vitro Antineoplastic Activity

Mocetinostat demonstrated potent antiproliferative activity across a diverse panel of human cancer cell lines, encompassing both hematologic malignancies and solid tumors.[11] Studies in breast cancer cell lines SUM149 and HCC1937 provided a clear mechanistic link between target engagement and cellular outcome. Treatment led to a dose-dependent increase in the acetylation of H3K9 and H3K27, confirming HDAC inhibition. This was followed by a marked suppression of cell proliferation, with

IC50​ values of 0.6 µM and 2.6 µM, respectively, and a reduction in colony-forming ability by approximately 80%.[10]

A key finding from these early studies was the apparent selectivity of Mocetinostat for malignant cells. Compared to the pan-HDAC inhibitor vorinostat, which exhibited strong antiproliferative effects against both cancer cells and normal human fibroblasts, Mocetinostat was found to be more potent against cancer cells while showing a lack of antiproliferative activity against normal fibroblasts.[11] This suggested a potentially superior therapeutic index and a more favorable safety profile, which was a significant driver for its clinical investigation.

In Vivo Efficacy

The anti-tumor activity observed in vitro was successfully translated to in vivo models. Mocetinostat effectively inhibited the growth of human tumor xenografts in mice, providing crucial proof-of-concept for its systemic anti-cancer efficacy.[11] These models also confirmed that the drug had good oral bioavailability, a critical property for its development as a convenient, orally administered therapy.[1]

Furthermore, preclinical studies were instrumental in guiding the design of rational combination therapies. A particularly important finding was the demonstration of in vitro and in vivo synergy between Mocetinostat and the hypomethylating agent 5-azacitidine.[27] These two classes of epigenetic drugs, acting on histone deacetylation and DNA methylation respectively, showed complementary and enhanced anti-leukemic activity. This preclinical evidence provided the direct and compelling rationale for initiating the Phase I/II clinical trial (NCT00324220) combining the two agents in patients with MDS and AML.[27] Pharmacodynamic studies in HCT116 human colon carcinoma cells helped to quantify the degree of target engagement, showing that the maximal inhibitable HDAC enzyme pool was approximately 75% of the total cellular activity, a plateau reached at a drug concentration of 6 µM.[16]

Section 5: Clinical Development and Efficacy Analysis

The clinical journey of Mocetinostat has been extensive, spanning numerous trials across a wide array of cancers and therapeutic combinations. This section provides a detailed, trial-by-trial analysis of its performance, revealing a pattern of clear biological activity often tempered by significant toxicity, and highlighting how its clinical utility is profoundly influenced by the choice of combination partner.

Trial IDPhaseIndication(s)RegimenNObjective Response Rate (ORR)Disease Control Rate (DCR)Key Conclusion/Status
NCT00359086IIDLBCL / Follicular LymphomaMocetinostat Monotherapy69DLBCL: 17%; FL: 11%DLBCL: 49%; FL: 61%Modest single-agent activity; further development in combinations warranted. 29
NCT00324220I/IIMDS / AMLMocetinostat + 5-Azacitidine20 (MDS)50% (CR+mCR in high-risk)80%Encouraging clinical benefit and acceptable safety; validates preclinical synergy. 27
NCT02236195IIUrothelial Carcinoma (CREBBP/EP300 mutated)Mocetinostat Monotherapy1711% (1 PR in 9 evaluable)N/AInsufficient activity; trial halted due to toxicity and low drug exposure. 30
NCT02805660I/IINon-Small Cell Lung CancerMocetinostat + Durvalumab8311.5% (Overall); 23.1% (in prior CPI refractory)N/ADurable activity in an immune-refractory population; tolerable combination. 31
NCT03565406IbUnresectable MelanomaMocetinostat + Nivolumab + Ipilimumab10~89% (in 70 mg cohort)N/AExceptionally high efficacy but prohibitive toxicity; trial terminated. 24
NCT02303262IIMetastatic LeiomyosarcomaMocetinostat + GemcitabineN/AN/AN/AFeasible, safe, and showed proof-of-principle activity in chemo-resistant disease. 34
N/AIRhabdomyosarcoma (R/R)Mocetinostat + Vinorelbine8N/A86% (Clinical Benefit Rate)Highly promising interim results with acceptable safety in a high-need population. 36

Application in Hematologic Malignancies

Relapsed/Refractory Lymphomas (NCT00359086)

This open-label, Phase II study evaluated Mocetinostat as a single agent in 69 patients with relapsed or refractory lymphoma, specifically Diffuse Large B-cell Lymphoma (DLBCL; n=41) and Follicular Lymphoma (FL; n=28).[29] Patients received Mocetinostat at doses ranging from 70-110 mg three times weekly. The study demonstrated modest but clear single-agent activity. In the heavily pretreated DLBCL cohort, the objective response rate (ORR) was 17%, with a disease control rate (DCR; defined as CR + PR + Stable Disease) of 49%. In the FL cohort, the ORR was 11%, which notably included one patient who achieved a durable complete response, and the DCR was 61%. While these response rates were not sufficient to position Mocetinostat as a transformative monotherapy, the evidence of clinical activity in these difficult-to-treat populations was considered a positive signal. The investigators concluded that the drug's acceptable tolerability and observed efficacy warranted further development, likely in combination with other agents to enhance its effect.[29]

Myelodysplastic Syndromes (MDS) and Acute Myeloid Leukemia (AML) (NCT00324220)

Building on strong preclinical data demonstrating synergy, this Phase I/II study evaluated Mocetinostat in combination with the hypomethylating agent 5-azacitidine (AZA) in patients with high-risk MDS or AML.[28] The MDS cohort (n=20) received standard-dose AZA (75 mg/m² for 7 days) with Mocetinostat (90-110 mg three times weekly).[27] The results from this cohort were highly encouraging and represented a significant clinical success for the drug. The combination yielded an impressive DCR of 80% (16 of 20 patients). In the highest-risk subgroup of patients with baseline marrow blast counts ≥10%, 50% achieved a complete response (CR) or marrow-CR. One notable case was a 74-year-old male with previously untreated disease whose marrow blasts fell from 11% to 0% after just one cycle, achieving a full CR that was maintained for an entire year on study. The combination demonstrated an acceptable safety profile, with most adverse events being Grade 1 or 2. This trial provided strong clinical validation for the rational, mechanism-based combination of two distinct epigenetic modifiers, demonstrating that Mocetinostat could be a highly effective partner for AZA in MDS.[27]

Application in Solid Tumors

Genomically-Selected Urothelial Carcinoma (NCT02236195)

This Phase II trial was a pioneering effort in precision oncology, designed to test Mocetinostat in a patient population with a strong biological hypothesis for sensitivity.[40] The study enrolled patients with platinum-pretreated, advanced urothelial carcinoma whose tumors harbored inactivating genetic alterations in the histone acetyltransferase (HAT) genes

CREBBP or EP300. The rationale was that tumors deficient in histone acetylation would be uniquely vulnerable to the inhibition of histone deacetylation. Despite this elegant design, the trial was a clinical failure and was halted after the first stage.[30]

Among nine evaluable patients, only one partial response was observed (ORR 11%), which was insufficient to meet the prespecified criteria for expansion. The trial's failure was not necessarily due to a flawed biological hypothesis but rather to the drug's challenging pharmaceutical properties. All patients experienced at least one adverse event, with nausea (77%) and fatigue (71%) being pervasive. These toxicities led to frequent treatment interruptions (in 82% of patients) and dose reductions (29%). Consequently, the median duration of treatment was only 46 days, and pharmacokinetic analysis revealed that drug exposure was lower than anticipated. This trial serves as a critical cautionary tale in the era of precision medicine: even the most compelling genomic rationale cannot overcome the fundamental challenges of a drug that is too toxic to be delivered consistently at a therapeutic dose.[30]

Advanced Non-Small Cell Lung Cancer (NSCLC) (NCT02805660)

This Phase I/II study provided the first major clinical test of Mocetinostat's immunomodulatory potential by combining it with durvalumab, an anti-PD-L1 checkpoint inhibitor.[31] The study enrolled 83 patients with advanced NSCLC, including cohorts of patients who had previously been treated with and progressed on checkpoint inhibitors. The recommended Phase II dose (RP2D) was established as Mocetinostat 70 mg three times weekly plus standard-dose durvalumab. The combination was generally well-tolerated, with fatigue (41%), nausea (40%), and diarrhea (31%) being the most common treatment-related adverse events.

The efficacy results provided important clinical proof-of-concept. The overall ORR across the Phase II cohorts was 11.5%, and responses were notably durable, with a median duration of 329 days. The most significant finding, however, was in the cohort of patients whose disease was refractory to prior anti-PD-(L)1 therapy. In this difficult-to-treat, immune-resistant population, the combination achieved an ORR of 23.1%. This result suggested that Mocetinostat could successfully re-sensitize tumors to checkpoint blockade, lending clinical credence to the preclinical data showing its ability to remodel the tumor microenvironment.[31]

Unresectable Melanoma (NCT03565406)

This Phase Ib pilot trial represented the most aggressive test of Mocetinostat's immunomodulatory synergy, combining it in a triplet regimen with dual checkpoint blockade: nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4) in 10 treatment-naïve patients with metastatic melanoma.[23] The results were a dramatic illustration of Mocetinostat's dual nature of promise and peril. The efficacy was extraordinary: in the cohort of nine patients treated with the 70 mg dose of Mocetinostat, eight achieved an objective response (one CR, seven PRs), for an ORR of approximately 89%.[33] These responses were also durable, with a median follow-up of over 16 months.[23]

However, this profound efficacy came at the cost of unacceptable toxicity. All ten patients treated experienced Grade 2 or higher adverse events, and a staggering seven patients had Grade 3-4 immune-related adverse events.[23] The severity of the toxicity profile led to the trial's termination.[32] This study powerfully demonstrated that Mocetinostat could synergize with dual checkpoint blockade to produce unprecedented response rates, but the combination amplified toxicity to a clinically untenable degree. It underscored the critical challenge of uncoupling the drug's potent efficacy from its severe adverse effects.[24]

Metastatic Leiomyosarcoma (NCT02303262)

This Phase II study explored a different combination strategy: using Mocetinostat to overcome chemotherapy resistance.[34] Patients with metastatic leiomyosarcoma who had previously progressed on gemcitabine-containing therapy were treated with a combination of Mocetinostat and gemcitabine. The trial successfully demonstrated the feasibility and safety of this combination and provided proof-of-principle for its activity. This suggests a potential role for Mocetinostat in re-sensitizing tumors to conventional cytotoxic agents, a valuable strategy for extending the benefit of established chemotherapies.[35]

Refractory/Recurrent Rhabdomyosarcoma (RMS)

An investigator-initiated Phase I dose-escalation and expansion trial has been evaluating Mocetinostat in combination with the chemotherapy agent vinorelbine in heavily pretreated adolescents and young adults with refractory or recurrent (R/R) rhabdomyosarcoma.[36] Interim results from the first eight patients have been exceptionally promising. In seven evaluable patients, the clinical benefit rate (CR+PR+SD) was 86%, with four patients achieving a partial response. Responses were both rapid, occurring at a median of 1.5 months, and durable, with a median duration of 8 months. Importantly, the combination has shown an acceptable safety profile. The primary Grade 3/4 toxicities were hematologic (neutropenia, lymphopenia, anemia), which were transient, reversible, and manageable with growth factor support. These early results are highly encouraging, suggesting that the combination of Mocetinostat and vinorelbine may represent a therapeutic "sweet spot"—a regimen that achieves powerful synergy and high efficacy with a manageable safety profile in a patient population with a profound unmet medical need.[36]

Section 6: Safety, Tolerability, and Risk Management

A comprehensive understanding of Mocetinostat's safety profile is critical to interpreting its clinical trial results and understanding the challenges that have shaped its development. A consistent pattern of adverse events has emerged across its extensive clinical program, alongside specific, significant risks related to drug interactions.

Adverse Event Profile

The most frequently reported treatment-related adverse events (AEs) associated with Mocetinostat are primarily constitutional and gastrointestinal in nature. Across multiple studies, including those in lymphoma, MDS, NSCLC, and urothelial carcinoma, the most common AEs include:

  • Fatigue [16]
  • Nausea [16]
  • Diarrhea [16]
  • Vomiting [16]
  • Anorexia or weight loss [16]

When examining more severe (Grade 3 or 4) events, a similar pattern holds, with nausea (15%), vomiting (10%), and fatigue (25% as a DLT in one study) being common.[16] In addition, clinically significant hematologic toxicities are frequently observed, particularly in combination regimens, including anemia, thrombocytopenia, and febrile neutropenia (10% each in the MDS study).[27] The safety profile can be significantly altered by the combination partner. For example, in the triplet therapy trial in melanoma (NCT03565406), severe immune-related adverse events were predominant and dose-limiting.[23]

Dose-Limiting Toxicities (DLTs) and Tolerability

The tolerability of Mocetinostat has been a persistent challenge throughout its clinical development. In early-phase dose-escalation studies, dose-limiting toxicities (DLTs) included nausea, vomiting, abdominal pain, deep vein thrombosis, diarrhea, and fatigue.[16] The maximum tolerated dose (MTD) was established at 90 mg three times per day in one study.[16]

The impact of this toxicity profile on treatment delivery is significant. In the single-agent lymphoma study (NCT00359086), 28% of patients had to discontinue the study drug due to adverse events.[29] Similarly, in the urothelial carcinoma trial (NCT02236195), the high rates of nausea and fatigue led to frequent treatment interruptions and dose reductions, which ultimately compromised drug exposure and likely contributed to the trial's failure.[30] This demonstrates that the AE profile is not just a matter of patient comfort but a critical determinant of the drug's ability to be administered at a therapeutically effective dose.

Significant Drug Interaction Risks

Beyond its intrinsic toxicity, Mocetinostat carries two major, well-documented risks for serious adverse events when combined with other medications. Clinical trial protocols have often included specific exclusion criteria to mitigate these risks, such as excluding patients with pre-existing cardiac abnormalities.[42]

QTc Prolongation

Mocetinostat has the potential to prolong the QTc interval on an electrocardiogram, which is a marker for delayed ventricular repolarization and a known risk factor for life-threatening cardiac arrhythmias like Torsades de Pointes. This risk is significantly increased when Mocetinostat is co-administered with a large number of other drugs that also share this property.[5] The list of interacting drugs is extensive and spans many common classes, including:

  • Antiarrhythmics: Amiodarone, Bepridil
  • Antipsychotics: Amisulpride, Asenapine
  • Antidepressants: Amitriptyline
  • Antibiotics: Ciprofloxacin, Azithromycin
  • Antihistamines: Astemizole, Bilastine
  • Other Agents: Amantadine, Atazanavir, Cilostazol, Buserelin (Representative examples from 5)

Methemoglobinemia

Mocetinostat can increase the risk or severity of methemoglobinemia, a rare but serious blood disorder in which an abnormal amount of methemoglobin—a form of hemoglobin that cannot bind and release oxygen—is produced.[5] This risk is particularly pronounced when Mocetinostat is combined with certain drugs, most notably local anesthetics. Clinicians should be aware of this potential interaction with agents such as:

  • Benzocaine
  • Lidocaine
  • Bupivacaine
  • Cinchocaine
  • Chloroprocaine
  • Articaine Other interacting agents include ambroxol and benzyl alcohol.5

Section 7: Regulatory Status and Designations

The regulatory history of an investigational drug provides valuable context for its development pathway, reflecting both early promise and subsequent strategic shifts by its sponsor. Mocetinostat has received orphan drug designations from both the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA), although several of these have since been withdrawn.

U.S. Food and Drug Administration (FDA)

The FDA granted Mocetinostat Orphan Drug Designation for two separate hematologic malignancies, a status that provides incentives such as market exclusivity and tax credits to encourage the development of drugs for rare diseases.[43]

  • Myelodysplastic Syndrome (MDS): Mocetinostat received Orphan Drug Designation for the treatment of MDS on June 12, 2014.[16] This designation was later officially withdrawn by the sponsor on March 21, 2022.[44]
  • Diffuse Large B-Cell Lymphoma (DLBCL): The FDA granted a second Orphan Drug Designation for the treatment of DLBCL in August 2014.[16]

European Medicines Agency (EMA)

Similarly, the EMA granted an orphan designation for Mocetinostat in the European Union.

  • Acute Myeloid Leukemia (AML): Mocetinostat (under its chemical name) was granted orphan designation (EU/3/07/526) for the treatment of AML on January 31, 2008.[16] This designation was also subsequently withdrawn at the sponsor's request in May 2022.[47]

The initial granting of these designations between 2008 and 2014 indicates a period of significant optimism and a clear development strategy focused on hematologic malignancies, where the drug had shown promising preclinical and early clinical signals. However, the formal withdrawal of the MDS and AML designations in 2022 is a powerful non-clinical indicator of a major strategic re-evaluation. A sponsor would not voluntarily relinquish the valuable benefits of an orphan designation for a high-priority asset. This action, therefore, strongly suggests that the development of Mocetinostat for these specific indications was de-prioritized or formally discontinued. This decision was likely influenced by the totality of the clinical data gathered over the subsequent years, including the challenging risk-benefit profile observed across the entire program, which may have led the sponsor, Mirati Therapeutics, to shift its focus and resources toward other assets in its pipeline.

Section 8: Integrated Analysis and Future Outlook

The extensive development history of Mocetinostat provides a rich and nuanced case study in modern oncology drug development. It is a molecule of clear and potent biological activity, yet its path has been fraught with challenges that have so far prevented its approval. A holistic analysis reveals a drug defined by a central tension between its therapeutic promise and the peril of its toxicity, with its ultimate clinical value appearing to be entirely dependent on the context of its use.

The Promise and Peril of Mocetinostat

The "promise" of Mocetinostat is multifaceted and undeniable. Its isotype-selective mechanism of action is well-defined, and it has consistently demonstrated the ability to induce desired cellular effects, including apoptosis and cell cycle arrest. More importantly, it possesses a potent, dual-pronged mechanism that combines direct cytotoxicity with profound immunomodulation. This has translated into compelling signals of clinical efficacy in several settings. The 80% disease control rate seen in high-risk MDS when combined with 5-azacitidine, the ~89% objective response rate in melanoma when combined with dual checkpoint blockade, and the 86% clinical benefit rate in heavily pretreated rhabdomyosarcoma when combined with vinorelbine all point to a drug capable of achieving powerful synergistic effects.

However, this promise is inextricably linked to its "peril"—a consistent and often dose-limiting toxicity profile dominated by gastrointestinal and constitutional symptoms. This toxicity has directly led to the failure of at least one clinical trial (in urothelial carcinoma) by preventing adequate drug exposure. In its most effective combination (in melanoma), it amplified toxicity to an unacceptable level, rendering the regimen clinically untenable. This difficult therapeutic window has been the primary obstacle to its successful clinical translation, forcing a continuous search for a context where its efficacy can be realized without prohibitive adverse events.

The Future of Isotype-Selective HDAC Inhibition

The experience with Mocetinostat raises important questions for the entire class of isotype-selective HDAC inhibitors. It is unclear whether the observed toxicities are idiosyncratic to the Mocetinostat molecule itself—perhaps due to off-target effects or specific pharmacokinetic properties—or if they represent an inherent on-target toxicity associated with the potent inhibition of HDACs 1, 2, 3, and 11. If the latter is true, it suggests that achieving a sufficiently wide therapeutic window with this specific target profile may be fundamentally challenging. The future of this field will likely depend on the development of next-generation inhibitors with even greater isoform selectivity (e.g., targeting only HDAC1/2) or novel chemical scaffolds with more favorable pharmacological properties that might successfully uncouple the desired epigenetic and immunomodulatory effects from dose-limiting toxicities.

Unanswered Questions and Avenues for Future Research

The story of Mocetinostat is not necessarily over, but its future path, and that of similar agents, will depend on addressing several critical questions:

  • Can Efficacy Be Uncoupled from Toxicity? The most pressing question is whether the therapeutic index can be improved. This could be explored through alternative dosing schedules, such as lower or less frequent dosing, designed to maintain the immunomodulatory effects (which may occur at lower concentrations) while mitigating the acute toxicities. The de-escalation to a 50 mg dose in the melanoma trial was a step in this direction, though that trial was terminated before this could be fully explored.
  • Identifying the Optimal Combination Partner: The clinical data strongly suggest that Mocetinostat is a combination agent, not a monotherapy. Its profile changes dramatically depending on its partner. While the combination with dual checkpoint blockade was too toxic, the combination with single-agent anti-PD-L1 in NSCLC was tolerable, and the combination with vinorelbine in rhabdomyosarcoma appears highly promising. Future success hinges on identifying more of these "sweet spot" combinations where synergy is maximized and toxicity is manageable.
  • Discovering Robust Predictive Biomarkers: The failure of the biomarker-driven urothelial cancer trial highlights the need for better predictors of response. While targeting CREBBP/EP300 mutations was a rational approach, it was not successful. Future studies should investigate whether biomarkers related to the tumor immune microenvironment—such as baseline levels of Tregs, MDSCs, or PD-L1 expression—might be more predictive of benefit, particularly for immunotherapy combinations.
  • Exploring Non-Oncological Indications: The promising preclinical data in cardiorenal protection and osteoarthritis should not be overlooked. It is plausible that Mocetinostat could be repurposed for these chronic, non-malignant conditions. Such indications might require much lower, better-tolerated doses administered over long periods, potentially offering a viable alternative development path where the drug's challenging toxicity profile in oncology is less of an impediment.

Works cited

  1. Mocetinostat (MGCD0103): a review of an isotype-specific histone deacetylase inhibitor, accessed September 9, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5206967/
  2. Histone acetyltransferases and histone deacetylases in B- and T-cell development, physiology and malignancy - Genes & Cancer, accessed September 9, 2025, https://www.genesandcancer.com/article/65/text/
  3. The Roles of Histone Deacetylases and Their Inhibitors in Cancer Therapy - Frontiers, accessed September 9, 2025, https://www.frontiersin.org/journals/cell-and-developmental-biology/articles/10.3389/fcell.2020.576946/full
  4. HDAC Inhibitors in Cancer Care, accessed September 9, 2025, https://www.cancernetwork.com/view/hdac-inhibitors-cancer-care
  5. Mocetinostat: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed September 9, 2025, https://go.drugbank.com/drugs/DB11830
  6. Roles of Histone Deacetylases and Inhibitors in Anticancer Therapy - MDPI, accessed September 9, 2025, https://www.mdpi.com/2072-6694/12/6/1664
  7. Synergistic Enhancement of Cancer Therapy Using ... - Frontiers, accessed September 9, 2025, https://www.frontiersin.org/journals/genetics/articles/10.3389/fgene.2020.578011/full
  8. Mocetinostat | C23H20N6O | CID 9865515 - PubChem, accessed September 9, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Mocetinostat
  9. Mocetinostat - Wikipedia, accessed September 9, 2025, https://en.wikipedia.org/wiki/Mocetinostat
  10. Mocetinostat | MGCD-0103 | CAS#726169-73-9 | 944537-89-7 - MedKoo Biosciences, accessed September 9, 2025, https://www.medkoo.com/products/5656
  11. Mocetinostat – Knowledge and References - Taylor & Francis, accessed September 9, 2025, https://taylorandfrancis.com/knowledge/Medicine_and_healthcare/Pharmaceutical_medicine/Mocetinostat/
  12. en.wikipedia.org, accessed September 9, 2025, https://en.wikipedia.org/wiki/Mocetinostat#:~:text=Mocetinostat%20(MGCD0103)%20is%20a%20benzamide,lymphoma%20and%20acute%20myelogenous%20leukemia.
  13. Mocetinostat (MGCD0103) | HDAC inhibitor | 99.93%(HPLC) | In Stock - Selleck Chemicals, accessed September 9, 2025, https://www.selleckchem.com/products/MGCD0103(Mocetinostat).html
  14. MGCD0103 ≥95% (HPLC) - Sigma-Aldrich, accessed September 9, 2025, https://www.sigmaaldrich.com/US/en/product/sigma/sml3453
  15. Mocetinostat (​MGCD0103, CAS Number: 726169-73-9) | Cayman Chemical, accessed September 9, 2025, https://www.caymanchem.com/product/18287/mocetinostat
  16. MOCETINOSTAT - Inxight Drugs, accessed September 9, 2025, https://drugs.ncats.io/drug/A6GWB8T96J
  17. Histone Deacetylase Inhibitors for Peripheral T-Cell Lymphomas - MDPI, accessed September 9, 2025, https://www.mdpi.com/2072-6694/16/19/3359
  18. Revisiting Histone Deacetylases in Human Tumorigenesis: The ..., accessed September 9, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6471041/
  19. Novel Role of Histone Deacetylase 11 (HDAC11) in Hematopoiesis | Request PDF, accessed September 9, 2025, https://www.researchgate.net/publication/336548539_Novel_Role_of_Histone_Deacetylase_11_HDAC11_in_Hematopoiesis
  20. HDAC Inhibitors in Acute Myeloid Leukemia - MDPI, accessed September 9, 2025, https://www.mdpi.com/2072-6694/11/11/1794
  21. The class I/IV HDAC inhibitor mocetinostat increases tumor antigen presentation, decreases immune suppressive cell types and augments checkpoint inhibitor therapy - PubMed, accessed September 9, 2025, https://pubmed.ncbi.nlm.nih.gov/29124315/
  22. The class I/IV HDAC inhibitor mocetinostat increases tumor antigen presentation, decreases immune suppressive cell types and augments checkpoint inhibitor therapy - ResearchGate, accessed September 9, 2025, https://www.researchgate.net/publication/320971489_The_class_IIV_HDAC_inhibitor_mocetinostat_increases_tumor_antigen_presentation_decreases_immune_suppressive_cell_types_and_augments_checkpoint_inhibitor_therapy
  23. Preclinical and clinical studies of a class I/IV HDAC inhibitor, mocetinostat, in melanoma., accessed September 9, 2025, https://ascopubs.org/doi/10.1200/JCO.2020.38.15_suppl.10052
  24. Clinical and immune correlate results from a phase 1b study of the histone deacetylase inhibitor mocetinostat with ipilimumab and nivolumab in unresectable stage III/IV melanoma | Request PDF - ResearchGate, accessed September 9, 2025, https://www.researchgate.net/publication/361202223_Clinical_and_immune_correlate_results_from_a_phase_1b_study_of_the_histone_deacetylase_inhibitor_mocetinostat_with_ipilimumab_and_nivolumab_in_unresectable_stage_IIIIV_melanoma
  25. Histone deacetylase inhibitor, mocetinostat, regulates cardiac remodelling and renin-angiotensin system activity in rats with transverse aortic constriction-induced pressure overload cardiac hypertrophy - IMR Press, accessed September 9, 2025, https://www.imrpress.com/journal/RCM/22/3/10.31083/j.rcm2203113/htm
  26. Mocetinostat activates Krüppel-like factor 4 and protects against tissue destruction and inflammation in osteoarthritis - JCI Insight, accessed September 9, 2025, https://insight.jci.org/articles/view/170513
  27. A phase II study of mocetinostat, an oral isotype-selective histone deacetylase (HDAC) inhibitor, in combination with 5-azacitidine in patients with myelodysplastic syndrome (MDS). - ASCO, accessed September 9, 2025, https://www.asco.org/abstracts-presentations/ABSTRACT117191
  28. A phase II study of mocetinostat, an oral isotype-selective histone deacetylase (HDAC) inhibitor, in combination with 5-azacitidine in patients with myelodysplastic syndrome (MDS). - ASCO Publications, accessed September 9, 2025, https://ascopubs.org/doi/10.1200/jco.2013.31.15_suppl.7116
  29. A phase II study of single agent mocetinostat, an oral isotype ... - ASCO, accessed September 9, 2025, https://www.asco.org/abstracts-presentations/ABSTRACT117882
  30. Mocetinostat for patients with previously treated, locally advanced ..., accessed September 9, 2025, https://pubmed.ncbi.nlm.nih.gov/30570744/
  31. Mocetinostat in Combination With Durvalumab for Patients With ..., accessed September 9, 2025, https://pubmed.ncbi.nlm.nih.gov/36890020/
  32. Mocetinostat Terminated Phase 1 Trials for Melanoma Treatment | DrugBank Online, accessed September 9, 2025, https://go.drugbank.com/drugs/DB11830/clinical_trials?conditions=DBCOND0020306&phase=1&purpose=treatment&status=terminated
  33. Clinical and Immune Correlate Results from a Phase 1b Study of the HDAC Inhibitor Mocetinostat with Ipilimumab and Nivolumab in Unresectable Stage III/IV Melanoma, accessed September 9, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9444873/
  34. Mocetinostat Completed Phase 2 Trials for Metastatic Leiomyosarcoma Treatment, accessed September 9, 2025, https://go.drugbank.com/drugs/DB11830/clinical_trials?conditions=DBCOND0075427&phase=2&purpose=treatment&status=completed
  35. SARC018-SPORE01 - SARC SARCOMA CLINICAL TRIALS, accessed September 9, 2025, https://sarctrials.org/clinical-trials/sarc018-spore01/
  36. A phase 1 dose-escalation/expansion clinical trial of mocetinostat in combination with vinorelbine in adolescents and young adults with refractory and/or recurrent rhabdomyosarcoma: Interim results. - ASCO Publications, accessed September 9, 2025, https://ascopubs.org/doi/10.1200/JCO.2022.40.16_suppl.11553
  37. Mocetinostat Completed Phase 2 Trials for Lymphoma Treatment | DrugBank Online, accessed September 9, 2025, https://go.drugbank.com/drugs/DB11830/clinical_trials?conditions=DBCOND0006647&phase=2&purpose=treatment&status=completed
  38. Mocetinostat Completed Phase 1 / 2 Trials for Acute Myeloid Leukemia / Myelodysplastic Syndrome Treatment | DrugBank Online, accessed September 9, 2025, https://go.drugbank.com/drugs/DB11830/clinical_trials?conditions=DBCOND0029883%2CDBCOND0028333&phase=1%2C2&purpose=treatment&status=completed
  39. NCT00324220 | A Phase I/II Study of MGCD0103 With Azacitidine in Patients With High-Risk Myelodysplastic Syndrome (MDS) or Acute Myelogenous Leukemia | ClinicalTrials.gov, accessed September 9, 2025, https://www.clinicaltrials.gov/study/NCT00324220?term=AREA%5BBasicSearch%5D(AREA%5BInterventionSearch%5D(MGCD-0103)%20AND%20AREA%5BStudyType%5D(INTERVENTIONAL))&rank=3
  40. An Open-Label, Single-Arm, Phase 2 Study of Mocetinostat in Selected Patients with Inactivating Alterations of Acetyltransferace Genes in Previously Treated Locally Advanced or Metastatic Urothelial Carcinoma. | Dana-Farber Cancer Institute, accessed September 9, 2025, https://www.dana-farber.org/clinical-trials/14-497
  41. Clinical and immune correlate results from a phase 1b study of the histone deacetylase inhibitor mocetinostat with ipilimumab and nivolumab in unresectable stage III/IV melanoma - PubMed, accessed September 9, 2025, https://pubmed.ncbi.nlm.nih.gov/35678233/
  42. Safety Study of Mocetinostat in Combination With Azacitidine in Subjects With MDS - Mayo Clinic, accessed September 9, 2025, https://www.mayo.edu/research/clinical-trials/cls-20111697
  43. Mirati Therapeutics Receives Orphan Designation from U.S. Food & Drug Administration for Mocetinostat in Diffuse Large B-Cell Lymphoma - PR Newswire, accessed September 9, 2025, https://www.prnewswire.com/news-releases/mirati-therapeutics-receives-orphan-designation-from-us-food--drug-administration-for-mocetinostat-in-diffuse-large-b-cell-lymphoma-270737161.html
  44. Search Orphan Drug Designations and Approvals - FDA, accessed September 9, 2025, https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=435414
  45. Mirati Therapeutics Receives Orphan Designation from U.S. Food & Drug Administration for Mocetinostat in Diffuse Large B-Cell Lymphoma - Pappas Capital, accessed September 9, 2025, https://www.pappas-capital.com/mirati-therapeutics-receives-orphan-designation-from-u-s-food-drug-administration-for-mocetinostat-in-diffuse-large-b-cell-lymphoma/
  46. Mirati's Mocetinostat For Lymphoma Designated As Orphan Drug - Bioprocess Online, accessed September 9, 2025, https://www.bioprocessonline.com/doc/mirati-s-mocetinostat-for-lymphoma-designated-as-orphan-drug-0001
  47. EU/3/07/526 - orphan designation for treatment of acute myeloid leukaemia, accessed September 9, 2025, https://www.ema.europa.eu/en/medicines/human/orphan-designations/eu-3-07-526

Published at: September 9, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.