Small Molecule
C26H34ClN3O4
1809336-39-7
Valemetostat, marketed under the brand name Ezharmia®, represents a significant advancement in the field of epigenetic therapy for cancer. It is a first-in-class, orally bioavailable, small molecule that functions as a dual inhibitor of the histone lysine methyltransferases Enhancer of Zeste Homolog 1 (EZH1) and Enhancer of Zeste Homolog 2 (EZH2).[1] These enzymes are the catalytic subunits of the Polycomb Repressive Complex 2 (PRC2), which plays a critical role in gene silencing. The dual-inhibition strategy is a key therapeutic innovation designed to overcome the compensatory activity of EZH1, a known mechanism of resistance that can limit the efficacy of selective EZH2 inhibitors.[2]
Developed by Daiichi Sankyo, Valemetostat has achieved landmark regulatory approvals from Japan's Pharmaceuticals and Medical Devices Agency (PMDA). In September 2022, it received its initial approval for the treatment of patients with relapsed or refractory (R/R) adult T-cell leukemia/lymphoma (ATL), a rare and aggressive hematologic malignancy with a high unmet medical need, particularly in Japan.[2] This was followed by an expanded approval in June 2024 for the treatment of adult patients with R/R peripheral T-cell lymphoma (PTCL), a heterogeneous group of aggressive non-Hodgkin lymphomas.[6]
The approvals were based on robust clinical data from pivotal Phase 2 trials. In the study for R/R ATL, Valemetostat demonstrated an impressive overall response rate (ORR) of 48.0% in a heavily pretreated population.[8] In the global VALENTINE-PTCL01 trial for R/R PTCL, the drug achieved a clinically meaningful ORR of 43.7% with a median duration of response (DOR) of nearly one year, a significant outcome in this difficult-to-treat setting.[10] The safety profile of Valemetostat is well-characterized and considered manageable. It is dominated by predictable, on-target hematologic toxicities, including thrombocytopenia, anemia, and neutropenia, which can typically be addressed with dose modifications and supportive care.[5]
Globally, Valemetostat remains an investigational agent. It has received Orphan Drug Designation (ODD) for the treatment of PTCL from both the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA), acknowledging the high unmet need in this rare disease.[13] The comprehensive data from its clinical development program, particularly the global VALENTINE-PTCL01 trial, will form the basis for potential regulatory submissions in these regions. This report provides an exhaustive analysis of Valemetostat, detailing its chemical properties, novel mechanism of action, preclinical evidence, pivotal clinical trial results, pharmacokinetic profile, and global regulatory journey, positioning it as a transformative therapy for specific hematologic malignancies.
A precise understanding of a drug's identity and fundamental chemical characteristics is essential for evaluating its pharmacological behavior, formulation, and clinical application. Valemetostat is a well-defined synthetic organic compound with a distinct set of identifiers and properties.
Valemetostat is recognized by several names and codes across scientific literature, clinical trial registries, and regulatory databases. The primary generic name is Valemetostat, which was assigned as its International Nonproprietary Name (INN).[1] In Japan, where it is commercially available, it is marketed under the brand name Ezharmia®.[2]
Throughout its development, it has been referred to by the code names DS-3201 and DS-3201b, which are frequently cited in early publications and clinical trial identifiers (e.g., NCT02732275).[1] Other synonyms include (R)-OR-S2.[16] The clinically developed and approved form is the tosylate salt, Valemetostat tosilate.[2] This distinction is important, as the salt form can significantly impact a drug's stability, solubility, and bioavailability.
For unambiguous identification in global databases, Valemetostat is assigned the DrugBank Accession Number DB18314 and the Chemical Abstracts Service (CAS) Number 1809336-39-7 for the base molecule.[1] The tosylate salt has a separate CAS Number, 1809336-93-3.[17] Other key identifiers include its PubChem Compound ID (CID) 126481870, FDA Unique Ingredient Identifier (UNII) 60RD0234VE, and KEGG ID D11551.[1]
Valemetostat is classified as a small molecule drug.[18] Its systematic International Union of Pure and Applied Chemistry (IUPAC) name is (2R)-7-chloro-2-[4-(dimethylamino)cyclohexyl]-N-[(4,6-dimethyl-2-oxo-1H-pyridin-3-yl)methyl]-2,4-dimethyl-1,3-benzodioxole-5-carboxamide.[1] This complex name describes a structure featuring several key functional groups, including a benzodioxole core, a substituted pyridinone moiety, and a dimethylamino-cyclohexyl group, which are critical for its binding and inhibitory activity.
The molecular formula of the base compound is C26H34ClN3O4, corresponding to a molecular weight of approximately 488.0 g/mol.[1] For computational chemistry and database searching, its structure can be represented by its SMILES (Simplified Molecular-Input Line-Entry System) and InChI (International Chemical Identifier) codes.[1]
Its physicochemical properties provide insight into its drug-like characteristics. Key computed descriptors include a LogP (a measure of lipophilicity) of approximately 4.4 (XLogP3), 2 hydrogen bond donors, and 5 hydrogen bond acceptors.[1] Its topological polar surface area (TPSA) is 79.9 Ų.[1] These values are generally within the ranges considered favorable for oral bioavailability. However, with a molecular weight approaching 500 Da and a LogP value that can exceed 5 in some calculations, it technically violates one of Lipinski's Rule of Five, a common characteristic of many modern, complex therapeutic agents designed for specific and potent target engagement.[20]
In terms of its physical state and solubility, Valemetostat is a crystalline solid that is insoluble in water but soluble in organic solvents such as dimethyl sulfoxide (DMSO) and ethanol.[16] This low aqueous solubility is a critical factor influencing its formulation for both preclinical and clinical use, necessitating specific delivery vehicles, such as carboxymethylcellulose sodium (CMC-Na) for oral suspensions in animal studies.[21]
Property | Value | Source(s) |
---|---|---|
Generic Name | Valemetostat | 1 |
Brand Name | Ezharmia® | 2 |
Developmental Code | DS-3201, DS-3201b | 1 |
DrugBank ID | DB18314 | 1 |
CAS Number (Base) | 1809336-39-7 | 1 |
CAS Number (Tosilate) | 1809336-93-3 | 17 |
IUPAC Name | (2R)-7-chloro-2-[4-(dimethylamino)cyclohexyl]-N-[(4,6-dimethyl-2-oxo-1H-pyridin-3-yl)methyl]-2,4-dimethyl-1,3-benzodioxole-5-carboxamide | 1 |
Molecular Formula | C26H34ClN3O4 | 1 |
Molecular Weight | 488.0 g/mol | 1 |
SMILES | CC1=CC(=C(C(=O)N1)CNC(=O)C2=CC(=C3C(=C2C)O[C@@](O3)(C)C4CCC(CC4)N(C)C)Cl)C | 1 |
InChIKey | SSDRNUPMYCFXGM-ZZHSESOFSA-N | 1 |
Solubility | Insoluble in water; Soluble in DMSO, Ethanol | 21 |
XLogP3 | 4.4 | 1 |
Hydrogen Bond Donors | 2 | 1 |
Hydrogen Bond Acceptors | 5 | 1 |
Polar Surface Area | 79.9 Ų | 1 |
The therapeutic strategy behind Valemetostat is rooted in a sophisticated understanding of cancer epigenetics, specifically targeting the dysregulation of histone methylation that drives many hematologic malignancies. Its mechanism as a dual inhibitor of EZH1 and EZH2 represents a rationally designed approach to achieve a more profound and durable anti-tumor effect than previously developed agents in its class.
Enhancer of Zeste Homolog 1 (EZH1) and Enhancer of Zeste Homolog 2 (EZH2) are histone-lysine N-methyltransferases that serve as the alternative catalytic subunits of the Polycomb Repressive Complex 2 (PRC2).[5] PRC2 is a master epigenetic regulator responsible for maintaining cellular identity and controlling gene expression programs during development and differentiation. Its primary enzymatic function is to catalyze the transfer of methyl groups to the lysine 27 residue of histone H3, leading to the formation of mono-, di-, and trimethylated H3K27 (H3K27me1/2/3).[18]
The H3K27me3 mark is a powerful repressive signal that leads to chromatin compaction, making the associated DNA inaccessible to the transcriptional machinery and thus silencing gene expression.[5] In a healthy cellular context, this process is tightly regulated to turn off genes that are not needed. However, in many cancers, particularly lymphomas and leukemias, the PRC2 pathway is hijacked. This often occurs through gain-of-function mutations in the
EZH2 gene or, more commonly in T-cell lymphomas, through the significant overexpression of the EZH2 protein.[1] This aberrant, hyperactive EZH2 leads to excessive H3K27me3 deposition across the genome, resulting in the inappropriate silencing of critical tumor suppressor genes. This epigenetic dysregulation blocks cellular differentiation, promotes uncontrolled proliferation, and is a key driver of oncogenesis.[1]
The discovery of EZH2's role as an oncogene led to the development of selective EZH2 inhibitors, such as tazemetostat. While these drugs have shown clinical activity, a significant limitation emerged from the biology of the PRC2 complex itself. EZH1, a close homolog of EZH2, can also function as the catalytic subunit of PRC2. Although EZH1 generally has weaker methyltransferase activity and is less abundant than EZH2 in many cell types, it can functionally compensate for the loss of EZH2 activity.[2]
When cancer cells are treated with a selective EZH2 inhibitor, EZH1 can be recruited to target gene promoters, where it continues to deposit the repressive H3K27me3 mark. This compensatory mechanism can sustain the oncogenic gene silencing program, thereby conferring intrinsic or acquired resistance to EZH2-selective therapy.[2]
Valemetostat was developed to overcome this fundamental challenge. By potently inhibiting both EZH1 and EZH2, it is designed to achieve a complete shutdown of PRC2-mediated methylation. This dual-inhibition strategy is not merely about increasing potency; it is a rational design to preemptively block a known biological escape route. Preclinical studies have validated this hypothesis, demonstrating that Valemetostat suppresses global H3K27me3 levels more profoundly and durably than selective EZH2 inhibitors. Crucially, Valemetostat treatment prevents the ectopic enrichment of EZH1 at tumor suppressor gene loci that is observed following treatment with tazemetostat.[2] This comprehensive blockade of the pathway leads to a more robust reactivation of silenced tumor suppressor genes, ultimately resulting in decreased cancer cell proliferation, induction of apoptosis, and cellular differentiation.[1]
This dual-action mechanism also suggests that Valemetostat's efficacy may be less dependent on the presence of specific EZH2 gain-of-function mutations. Selective inhibitors like tazemetostat show their greatest efficacy in follicular lymphoma patients with EZH2 mutations (ORR ~69%) compared to those with wild-type EZH2 (ORR ~35%).[27] In contrast, many T-cell lymphomas are driven by EZH2
overexpression rather than mutation.[3] In this context, where both EZH1 and EZH2 contribute to the oncogenic state, a dual inhibitor is theoretically more effective. This is supported by clinical findings that Valemetostat demonstrates activity in patients regardless of their
EZH2 mutation status, potentially broadening its clinical utility to a wider range of hematologic malignancies.[2]
Valemetostat acts as a potent, orally bioavailable, S-adenosylmethionine (SAM)-competitive inhibitor of both EZH1 and EZH2.[20] Its inhibitory activity has been quantified in cell-free enzymatic assays, which measure the direct interaction between the drug and its target enzymes. These assays have consistently shown potent, low-nanomolar inhibition of both homologs.
Reported half-maximal inhibitory concentration (IC50) values for Valemetostat are:
These data confirm that Valemetostat is a potent inhibitor of both targets, with a modest 3- to 4-fold selectivity for EZH2 over EZH1. The key therapeutic distinction lies in its nanomolar potency against EZH1, which is the basis of its dual-inhibitor classification.
Beyond enzymatic assays, Valemetostat has demonstrated exceptional potency within a cellular context. In HCT116 cancer cells, it inhibited the trimethylation of H3K27 with an IC50 of just 0.44 nM, indicating that it can effectively engage its target and modulate the epigenetic landscape at sub-nanomolar concentrations inside living cells.[16]
Inhibitor | EZH1 Potency (IC50 / Ki) | EZH2 Potency (IC50 / Ki) | Selectivity (EZH1/EZH2) | Source(s) |
---|---|---|---|---|
Valemetostat | 8.4 - 10.0 nM (IC50) | 2.5 - 6.0 nM (IC50) | ~3-4 fold for EZH2 | 2 |
Tazemetostat | 392 nM (IC50) | 11 nM (IC50); 2.5 nM (Ki) | ~35 fold for EZH2 | 29 |
Before advancing to human trials, Valemetostat underwent extensive preclinical evaluation to confirm its anti-cancer activity and validate its mechanism of action in relevant disease models. These studies provided the foundational evidence of its potential as a therapeutic agent for hematologic malignancies.
In cell-based assays, Valemetostat demonstrated potent and broad anti-proliferative effects across a range of hematological cancer cell lines. It consistently inhibited the growth of various non-Hodgkin lymphoma (NHL) cell lines with a half-maximal growth inhibition concentration (GI50) of less than 100 nM.[5] This activity was observed irrespective of the
EZH2 mutation status of the cells, supporting the hypothesis that its dual mechanism provides broader utility than selective inhibitors.[2]
The activity was specifically confirmed in cell lines derived from diseases that would later become its primary clinical targets. Valemetostat showed anti-proliferative effects against the TL-Om1 cell line, which is derived from human adult T-cell leukemia/lymphoma (ATL).[5] Furthermore, it was effective in models of diffuse large B-cell lymphoma (DLBCL), the most common type of NHL. It inhibited the growth of the Karpas-422 DLBCL cell line with a high degree of potency, showing a
GI50 of just 4.8 nM.[16]
The finding that Valemetostat is active against both the activated B-cell-like (ABC) and germinal center B-cell-like (GCB) subtypes of DLBCL is particularly noteworthy.[5] These two subtypes are biologically distinct, driven by different oncogenic pathways, and often respond differently to targeted therapies. For example, inhibitors of the B-cell receptor signaling pathway are primarily effective in the ABC subtype. The ability of Valemetostat to inhibit both subtypes suggests that the epigenetic dysregulation mediated by EZH1 and EZH2 is a fundamental dependency common to both forms of the disease. This provides a strong rationale for its clinical investigation in a broad population of B-cell lymphoma patients, as is being conducted in the ongoing VALYM trial.[3]
Mechanistically, Valemetostat was shown to induce apoptosis (programmed cell death) in DLBCL cell lines and to suppress the expression of BCL6, a master transcriptional repressor and a key oncogene in the pathogenesis of B-cell lymphomas.[5] This provides a direct link between the epigenetic reprogramming induced by Valemetostat and the downstream anti-tumor effects.
The promising in-vitro results were successfully translated into animal models of human cancer. Studies demonstrated that Valemetostat could not only block the survival of primary ATL cells in culture but also significantly reduce tumor growth in in-vivo xenograft models of both ATL and DLBCL.[5] These experiments were crucial in confirming that the drug could achieve sufficient exposure and target engagement in a living organism to produce a meaningful anti-tumor response, paving the way for its entry into clinical trials.
Further confirmation of its in-vivo target engagement came from a non-oncology study in mice, where administration of Valemetostat was shown to prevent the physiological changes in H3K27me3 levels that occur in skeletal muscle after exercise.[23] While the context is different, this study provided clear evidence of the drug's oral bioavailability and its ability to modulate its epigenetic target systemically.
The pharmacokinetic profile of Valemetostat, which describes its absorption, distribution, metabolism, and excretion (ADME), has been well-characterized through studies in healthy volunteers and patients with cancer. These properties are critical for determining the appropriate dosing regimen and understanding potential drug-drug interactions.
Valemetostat is formulated for oral administration and is rapidly absorbed into the systemic circulation following ingestion.[5] In clinical studies conducted under fasting conditions, the time to reach maximum plasma concentration (
Tmax) is typically between 2.0 and 4.5 hours.[33] The exposure to Valemetostat, as measured by its maximum concentration (
Cmax) and the area under the concentration-time curve (AUC), increases in a dose-proportional manner with single oral doses ranging from 50 mg to 200 mg.[33]
A clinically critical characteristic of Valemetostat's absorption is a significant negative food effect. The presence of food in the gastrointestinal tract drastically impairs its absorption, leading to a 50-60% reduction in Cmax and a 30-50% reduction in total drug exposure (AUC).[5] This reduction is substantial enough to potentially render the drug sub-therapeutic, making patient adherence to dosing instructions paramount for achieving clinical efficacy. Consequently, the approved labeling and clinical trial protocols strictly mandate that Valemetostat be administered on an empty stomach, defined as at least one hour before or two hours after a meal.[5] This requirement places a notable burden on patient education and compliance to ensure consistent and effective treatment in the real-world setting.
Once absorbed, Valemetostat is extensively distributed throughout the body. It is highly bound to plasma proteins, with over 94% of the drug bound at clinically relevant concentrations.[31] In-vitro studies have revealed that it binds with a higher affinity to alpha-1-acid glycoprotein (AAG) than to human serum albumin (HSA).[31] This is an important detail, as AAG is an acute-phase reactant protein whose levels can be elevated in patients with cancer or inflammatory conditions, which could potentially alter the fraction of unbound, active drug. Valemetostat shows minimal association with red blood cells, with a blood-to-plasma radioactivity ratio of 0.54, indicating that it primarily remains within the plasma compartment of the blood.[31]
The primary route of metabolism for Valemetostat is through oxidation by the cytochrome P450 (CYP) enzyme system in the liver. The main enzymes responsible are CYP3A4 and CYP3A5, with a minor contribution from CYP2C8.[33] This metabolic pathway leads to the formation of several metabolites, with the most abundant being a compound designated CALZ-1809a.[31] In addition to being a substrate for CYP enzymes, in-vitro studies have also identified Valemetostat as a substrate for the efflux transporter P-glycoprotein (P-gp), which can influence its absorption and distribution.[33]
Valemetostat and its metabolites are eliminated from the body primarily through the biliary/fecal route.[31] A human mass balance study using a single oral dose of radiolabeled [14C]-valemetostat provided a detailed account of its excretion pathways. Over a period of 360 hours, a mean of 95.3% of the administered radioactive dose was recovered. The majority of this, 79.8%, was found in the feces, while a smaller portion, 15.6%, was recovered in the urine.[31]
A significant portion of the drug is excreted unchanged. Unaltered Valemetostat accounted for 64.9% of the administered dose in the feces and 10.0% in the urine.[31] This indicates that while metabolism occurs, a substantial fraction of the absorbed dose is cleared from the body intact. The apparent terminal elimination half-life (
t1/2) of Valemetostat is approximately 20 hours, which is consistent with and supportive of a once-daily dosing schedule.[31]
Given its reliance on CYP3A and P-gp for metabolism and transport, Valemetostat has a significant potential for drug-drug interactions (DDIs). Clinical studies were conducted to quantify this risk. Co-administration with itraconazole, a strong inhibitor of both CYP3A and P-gp, resulted in a dramatic increase in Valemetostat exposure, with the Cmax increasing 2.9-fold and the AUC increasing 4.2-fold.[34] When administered with fluconazole, a moderate CYP3A inhibitor, Valemetostat
Cmax and AUC both increased by approximately 1.6-fold.[35] These findings confirm that concomitant use of strong or moderate CYP3A inhibitors can lead to clinically significant overexposure to Valemetostat, increasing the risk of toxicity. Therefore, dose reductions are warranted when Valemetostat must be used with such medications.[35]
The clinical development program for Valemetostat has successfully demonstrated its efficacy and established its role in treating specific, difficult-to-treat T-cell malignancies. The results from two pivotal Phase 2 trials were instrumental in securing its regulatory approvals in Japan and form the basis for its ongoing global development.
This multicenter, single-arm Phase 2 study was the first pivotal trial for Valemetostat and led to its initial marketing approval. The trial enrolled 25 Japanese patients with relapsed or refractory (R/R) aggressive subtypes of adult T-cell leukemia/lymphoma (ATL), a malignancy with a notoriously poor prognosis after first-line therapy.[9] The patient population was heavily pretreated, having received a median of three prior lines of therapy. Critically, 24 of the 25 patients (96%) had previously been treated with mogamulizumab, a standard-of-care therapy for ATL in Japan, indicating that the study population was refractory to established treatments.[9]
Patients received Valemetostat at a dose of 200 mg orally once daily.[9] The trial successfully met its primary endpoint, demonstrating a clinically and statistically significant anti-tumor effect.
Efficacy Metric | Result | Source(s) |
---|---|---|
Overall Response Rate (ORR) | 48.0% (12/25 patients) (90% CI: 30.5-65.9) | 8 |
Complete Response (CR) Rate | 20.0% (5/25 patients) | 8 |
Partial Response (PR) Rate | 28.0% (7/25 patients) | 8 |
Median Duration of Response (DOR) | Not Reached (95% CI: 1.87 months - NR) | 9 |
Median Progression-Free Survival (PFS) | 7.4 months | 9 |
Median Overall Survival (OS) | 16.4 months | 9 |
The results were highly encouraging. An ORR of 48.0% in this patient population represents a significant clinical benefit. The durability of these responses was particularly notable, with the median DOR not being reached at the time of the primary analysis.[37] The median overall survival of 16.4 months is a substantial improvement in a disease where survival after relapse is typically very short.[9]
Building on the success in ATL, the global VALENTINE-PTCL01 trial evaluated Valemetostat in a broader population of patients with various subtypes of R/R peripheral T-cell lymphoma (PTCL). This open-label, single-arm Phase 2 study enrolled 133 patients across Asia, Europe, North America, and Oceania who had received at least one prior systemic therapy.[6] The primary endpoint was the ORR as assessed by a blinded independent central review (BICR) based on CT imaging.[10]
The findings from this trial confirmed the strong and durable activity of Valemetostat in PTCL and supported its second approval in Japan.
Efficacy Metric | Result (n=119 efficacy-evaluable) | Source(s) |
---|---|---|
Overall Response Rate (ORR) | 43.7% (52/119 patients) (95% CI: 34.6-53.1) | 6 |
Complete Response (CR) Rate | 14.3% (17/119 patients) | 10 |
Partial Response (PR) Rate | 29.4% (35/119 patients) | 10 |
Median Duration of Response (DOR) | 11.9 months (95% CI: 7.8 - NE) | 10 |
Median Progression-Free Survival (PFS) | 5.5 months (95% CI: 3.5-8.3) | 11 |
Median Overall Survival (OS) | 17.0 months (95% CI: 13.5 - NE) | 11 |
In the aggressive R/R PTCL setting, where median OS after relapse is historically only around 5.8 months, the median DOR of nearly 12 months achieved with Valemetostat is a profound clinical outcome.[7] This durability suggests that targeting the fundamental epigenetic machinery with a dual EZH1/2 inhibitor may induce more stable disease control compared to therapies targeting signaling pathways that can be more easily bypassed by resistance mechanisms.
Furthermore, responses were observed across all major PTCL subtypes, with numerically higher response rates seen in subtypes with a T-follicular helper (TFH) phenotype, such as angioimmunoblastic T-cell lymphoma (AITL), where the ORR was 54.8%.[10] A particularly important finding from this trial was that 10 patients (8.4%) were able to use Valemetostat as a "bridge-to-transplant," achieving a sufficient response to become eligible for and proceed to a potentially curative allogeneic hematopoietic cell transplant (HCT).[12] For patients with R/R PTCL, allogeneic HCT offers the only chance for a long-term cure, but eligibility requires achieving a deep response to salvage therapy. The ability of Valemetostat to enable a meaningful proportion of patients to reach this critical milestone elevates its clinical role from a palliative therapy to a crucial component of a curative treatment strategy for a subset of patients.
The safety and tolerability of Valemetostat have been consistently characterized across its clinical development program. The adverse event profile is largely predictable and reflects the drug's mechanism of action, with myelosuppression being the most prominent on-target effect. The data from the large, global VALENTINE-PTCL01 trial (n=133) provides the most comprehensive overview of its safety profile.[10]
Valemetostat is generally considered to have a manageable safety profile. The majority of adverse events are hematologic and can be addressed through careful monitoring, dose modifications (interruptions or reductions), and standard supportive care measures like transfusions or growth factor support.[5] The prominent hematologic toxicities are a direct and expected consequence of the drug's mechanism. EZH1 and EZH2 are known to be essential for the normal function and maintenance of hematopoietic stem cells.[3] Therefore, systemic inhibition of these enzymes logically impacts hematopoiesis, leading to cytopenias. This understanding is critical for clinicians, as it frames these adverse events not as unexpected toxicities but as dose-dependent, on-target effects that can be proactively managed.
The most frequently reported TEAEs in the VALENTINE-PTCL01 study are summarized below.
Adverse Event | Any Grade (%) | Grade 3-4 (%) | Source(s) |
---|---|---|---|
Thrombocytopenia | 49.6 | 23.3 | 10 |
Anemia | 35.3 | 18.8 | 10 |
Diarrhea | 29.3 | 3.8 | 10 |
Dysgeusia (Altered Taste) | 28.6 | 0 | 10 |
Neutropenia | 26.3 | 17.3 | 10 |
COVID-19 | 21.1 | 3.0 | 10 |
Nausea | 17.3 | 0.8 | 10 |
Pyrexia (Fever) | 15.0 | 0 | 10 |
Cough | 15.0 | 0 | 10 |
While TEAEs were common, severe and life-threatening events were less frequent. In the VALENTINE-PTCL01 trial, serious TEAEs of any cause were reported in 39.8% of patients. However, serious adverse events considered by investigators to be related to Valemetostat treatment (serious TRAEs) occurred in only 6.8% of patients.[12]
TEAEs led to the death of 11.3% of patients in the trial, which is not unexpected in a population with advanced, aggressive cancer. Critically, however, no deaths were assessed as being related to Valemetostat treatment.[10] The rate of treatment discontinuation due to treatment-related adverse events was low, at 6.8%, indicating that most toxicities could be managed without stopping the therapy altogether.[10]
Proactive management through dose adjustments is a cornerstone of maintaining patients on therapy. In the VALENTINE-PTCL01 study, treatment-related adverse events led to dose interruptions in 31.6% of patients and dose reductions in 12.0%.[10] Thrombocytopenia was the most common reason for these modifications, underscoring the importance of regular blood count monitoring.[12]
Based on preclinical and clinical data, specific precautions are advised. Patients must be carefully monitored for myelosuppression throughout treatment.[5] Additionally, animal studies have reported evidence of embryofetal toxicity and teratogenicity at exposures relevant to the clinical dose. Therefore, effective contraception is required for patients of childbearing potential during treatment and for a defined period after the final dose.[5]
The regulatory journey of Valemetostat highlights a strategic, "Japan-first" approach by its developer, Daiichi Sankyo. By leveraging the higher prevalence of T-cell malignancies in Asia and utilizing Japan's expedited regulatory pathways, the company successfully brought the drug to its domestic market while continuing broader global development.
Valemetostat's development and approval process in Japan was accelerated by a key regulatory designation. In April 2019, Japan's Ministry of Health, Labour and Welfare (MHLW) granted Valemetostat the SAKIGAKE Designation for the treatment of R/R PTCL.[2] This system, analogous to the FDA's Breakthrough Therapy Designation, is designed to promote the research and development of innovative medicines for severe diseases and provides benefits such as prioritized consultation and a shortened review time (from 12 months to 6 months).[15]
This designation paved the way for its first marketing approval. On September 26, 2022, the MHLW approved Ezharmia® (valemetostat tosilate) for the treatment of patients with R/R ATL.[2] This was a landmark event, making Valemetostat the first dual EZH1/EZH2 inhibitor to be approved for therapeutic use anywhere in the world.[3]
Following the successful results of the global VALENTINE-PTCL01 trial, Daiichi Sankyo secured a second indication. In June 2024, the MHLW approved Ezharmia® for the treatment of adult patients with R/R PTCL.[6]
In the United States, Valemetostat remains an investigational drug and is not yet approved for any indication.[6] Recognizing the significant unmet need for new therapies in T-cell lymphomas, the FDA granted Valemetostat Orphan Drug Designation (ODD) for the treatment of PTCL on December 1, 2021.[3] The ODD program provides incentives, such as tax credits for clinical trials and potential market exclusivity, to encourage the development of drugs for rare diseases. The comprehensive data package from the VALENTINE-PTCL01 trial is expected to serve as the foundation for a future New Drug Application (NDA) submission to the FDA.[6]
The regulatory status in Europe mirrors that in the United States. Valemetostat is currently an investigational agent and does not have marketing authorization from the European Medicines Agency (EMA).[6] On February 24, 2022, the EMA granted Valemetostat an Orphan Designation for the treatment of peripheral T-cell lymphoma.[14] This designation provides scientific and regulatory support to facilitate its development. The global VALENTINE-PTCL01 trial included multiple sites within the European Union, and its results will be central to any future Marketing Authorisation Application (MAA) submitted to the EMA.[6]
Date | Regulatory Body | Milestone | Indication | Source(s) |
---|---|---|---|---|
April 2019 | PMDA (Japan) | SAKIGAKE Designation | Relapsed/Refractory Peripheral T-cell Lymphoma (PTCL) | 2 |
December 2021 | FDA (USA) | Orphan Drug Designation | Treatment of Peripheral T-cell lymphoma (PTCL) | 3 |
February 2022 | EMA (Europe) | Orphan Designation | Treatment of peripheral T-cell lymphoma | 14 |
September 2022 | PMDA (Japan) | Marketing Approval | Relapsed/Refractory Adult T-cell Leukemia/Lymphoma (ATL) | 2 |
June 2024 | PMDA (Japan) | Expanded Approval | Relapsed/Refractory Peripheral T-cell Lymphoma (PTCL) | 6 |
Valemetostat has successfully established itself as a pioneering epigenetic therapy, validating the dual EZH1/EZH2 inhibition strategy with two approvals in Japan. Its future trajectory will be defined by its ability to navigate a competitive landscape, expand into new indications, and secure global regulatory approvals.
The most direct and relevant competitor for Valemetostat is tazemetostat (Tazverik®), an FDA-approved selective EZH2 inhibitor. The key differentiator is Valemetostat's dual mechanism of action. This provides a compelling scientific rationale for superior efficacy by preventing the compensatory activity of EZH1, which can be a resistance mechanism to tazemetostat.[2] This mechanistic advantage may translate into more durable responses and broader activity in tumors that are not strictly dependent on an
EZH2 mutation, such as the T-cell lymphomas where Valemetostat has demonstrated its efficacy.[39] While tazemetostat is approved for follicular lymphoma (a B-cell malignancy) and epithelioid sarcoma, Valemetostat has strategically carved out a distinct clinical niche by first proving its value in T-cell malignancies, where the unmet need is particularly high.
The competitive landscape also includes emerging dual EZH1/2 inhibitors. At the 2023 ASH Annual Meeting, Haihe Biopharma presented data on its compound, HH2853, in PTCL. In a small, single-country (China) Phase 1b trial, HH2853 showed a numerically higher ORR of 65% compared to Valemetostat's 44% in the larger, global VALENTINE-PTCL01 trial.[44] While these results suggest a potential efficacy challenge, cross-trial comparisons must be interpreted with extreme caution. The VALENTINE-PTCL01 study was much larger and included a more geographically and ethnically diverse patient population, making its results more robust and generalizable. Nonetheless, the emergence of other dual inhibitors indicates that this will become a competitive therapeutic class.
The future growth for Valemetostat lies in expanding its use beyond the currently approved indications in Japan. Several key development programs are underway:
Valemetostat (Ezharmia®) is a landmark achievement in oncology drug development, representing the first-in-class dual inhibitor of EZH1 and EZH2 to gain regulatory approval. Its development was predicated on a sophisticated biological rationale: to overcome the compensatory resistance mechanisms that can limit the efficacy of selective EZH2 inhibitors. By comprehensively blocking PRC2-mediated gene silencing, Valemetostat has demonstrated the ability to induce deep and, most importantly, durable responses in patients with relapsed or refractory T-cell malignancies, a population with a historically grim prognosis and few effective treatment options.
The clinical data from pivotal trials in R/R ATL and R/R PTCL are compelling, showing clinically meaningful response rates and a median duration of response approaching or exceeding one year. A particularly significant outcome is its ability to serve as a bridge to potentially curative allogeneic stem cell transplantation for a subset of patients. Its safety profile, characterized by manageable, on-target myelosuppression, is well-understood and consistent with its mechanism of action.
Having secured two key approvals in Japan, Valemetostat has established a new standard of care and validated its therapeutic concept. The immediate future will focus on securing regulatory approvals in the United States and Europe, where it holds Orphan Drug Designation. Long-term success will depend on the outcomes of ongoing trials in B-cell lymphomas and solid tumors, its performance against emerging competitors, and the strategic development of rational combination therapies to further enhance its efficacy and overcome resistance. Valemetostat stands as a testament to mechanism-based drug design and holds significant promise to improve outcomes for a wide range of patients with hematologic and other cancers.
Published at: September 1, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.