MedPath

Pevonedistat Advanced Drug Monograph

Published:Aug 19, 2025

Generic Name

Pevonedistat

Drug Type

Small Molecule

Chemical Formula

C21H25N5O4S

CAS Number

905579-51-3

Pevonedistat (MLN4924): A Comprehensive Review of a First-in-Class NEDD8-Activating Enzyme Inhibitor from Preclinical Promise to Pivotal Trial Outcomes

Executive Summary

Pevonedistat (MLN4924) is an investigational, first-in-class small molecule inhibitor of the NEDD8-Activating Enzyme (NAE), a critical upstream regulator of the ubiquitin-proteasome system. By targeting the neddylation pathway, Pevonedistat disrupts the activity of Cullin-RING E3 ubiquitin ligases (CRLs), leading to the accumulation of specific CRL substrate proteins. This accumulation induces a state of proteotoxic stress characterized by cell cycle disruption, DNA damage, and ultimately, apoptosis in malignant cells. This novel mechanism of action positioned Pevonedistat as a promising therapeutic agent for hematologic malignancies, particularly for older, transplant-ineligible patients with higher-risk myelodysplastic syndromes (HR-MDS), chronic myelomonocytic leukemia (CMML), and acute myeloid leukemia (AML), where the standard of care has long been limited to hypomethylating agents (HMAs) with suboptimal outcomes.

The clinical development of Pevonedistat followed a trajectory of significant promise followed by profound disappointment. Early phase studies established a manageable safety profile and a recommended Phase 2 dose when combined with azacitidine. A randomized Phase 2 trial (NCT02610777) generated compelling evidence of clinical activity, particularly in the HR-MDS subgroup, where the combination of Pevonedistat and azacitidine nearly doubled the complete remission rate and significantly improved event-free survival compared to azacitidine alone. These highly encouraging results led the U.S. Food and Drug Administration (FDA) to grant Pevonedistat a Breakthrough Therapy Designation in 2020, signaling its potential to be the first novel treatment for this patient population in over a decade.

However, the subsequent global, randomized Phase 3 PANTHER trial (NCT03268954), designed to confirm these findings, failed to meet its primary endpoint of event-free survival. The statistically significant benefits observed in the Phase 2 HR-MDS cohort were not replicated in the larger, more definitive study. Analysis of this outcome suggests a multifactorial cause, including the unexpectedly strong performance of the azacitidine monotherapy control arm and the potential for patient heterogeneity to dilute the treatment effect. Critically, a post-hoc analysis revealed that patients who were able to remain on Pevonedistat combination therapy for an extended duration (e.g., more than six cycles) did experience a significant overall survival benefit, suggesting that the drug's efficacy may be conditional upon sustained treatment.

Following the Phase 3 trial failure, the sponsor withdrew the European Medicines Agency (EMA) orphan designations for Pevonedistat in both MDS and AML. Current research has pivoted to exploring alternative combination strategies, though a subsequent Phase 2 trial of a triplet regimen with venetoclax and azacitidine also failed to show a benefit over the doublet. The story of Pevonedistat serves as a critical case study in the complexities of oncology drug development, highlighting the challenges of translating Phase 2 signals to Phase 3 success and underscoring the importance of trial design, control arm optimization, and the potential need for biomarker-driven patient selection to unlock the potential of targeted therapies.

1.0 Introduction: Targeting the Neddylation Pathway in Hematologic Malignancies

1.1 The Unmet Need in Higher-Risk Myelodysplastic Syndromes (HR-MDS) and Acute Myeloid Leukemia (AML)

Higher-risk myelodysplastic syndromes (HR-MDS), chronic myelomonocytic leukemia (CMML), and acute myeloid leukemia (AML) with 20-30% bone marrow blasts represent a continuum of aggressive, clonal hematopoietic stem cell neoplasms.[1] These diseases are characterized by ineffective hematopoiesis, leading to cytopenias, and a significant risk of transformation to overt AML.[2] They predominantly affect older adults, with a median age at diagnosis of approximately 70 to 75 years, an age group often burdened with comorbidities that preclude the use of intensive, potentially curative therapies like allogeneic stem cell transplantation.[1]

For these transplant-ineligible patients, the therapeutic landscape has been stagnant for over a decade.[5] The standard of care has been treatment with hypomethylating agents (HMAs), such as azacitidine or decitabine.[7] While HMAs have demonstrated a modest survival advantage over conventional care regimens, the overall prognosis for patients with HR-MDS remains poor, with a median survival of only 1 to 3 years.[4] The majority of patients do not achieve a complete remission, and virtually all will eventually experience disease progression or relapse.[1] This clinical reality establishes a clear and urgent unmet medical need for novel therapeutic agents that can be safely combined with the HMA backbone to improve the depth and durability of response and extend survival in this vulnerable patient population.[11]

1.2 Pevonedistat: A Novel Therapeutic Approach

In response to this need, Pevonedistat (also known as MLN4924) was developed as a first-in-class, investigational small-molecule inhibitor of the NEDD8-Activating Enzyme (NAE).[2] The rationale for this approach stems from the critical role of the neddylation pathway in cellular homeostasis and its frequent dysregulation in cancer.[15] Neddylation is a post-translational modification process, analogous to ubiquitination, that is essential for the function of the largest family of E3 ubiquitin ligases, the Cullin-RING ligases (CRLs).[11] Upregulation of this pathway is associated with the pathogenesis of various malignancies, making NAE a compelling therapeutic target.[15] The therapeutic hypothesis for Pevonedistat was that by inhibiting NAE, the foundational enzyme of the neddylation cascade, it could disrupt CRL-mediated protein turnover, leading to the accumulation of tumor-suppressive proteins and ultimately inducing cancer cell death.[2]

The development of Pevonedistat represents a strategic evolution in targeting the ubiquitin-proteasome system (UPS) for cancer therapy. For years, the field has leveraged global proteasome inhibitors, such as bortezomib, which block the final step of protein degradation.[11] While effective, this approach can be associated with significant off-target effects and toxicities due to its indiscriminate blockade of the cell's primary protein disposal machinery. Pevonedistat offers a more refined, upstream intervention. Instead of inhibiting the proteasome directly, it selectively prevents the neddylation-dependent activation of CRLs.[15] This was theorized to disrupt the turnover of only a specific subset of proteins regulated by CRLs, potentially offering a more favorable therapeutic window and a distinct, more manageable toxicity profile.[11] This targeted approach was particularly appealing for the older, often frail HR-MDS and AML patient populations, for whom tolerability is a paramount concern.

2.0 Molecular Profile and Physicochemical Properties

2.1 Chemical Identity and Structure

Pevonedistat is a synthetic small molecule that has been assigned multiple identifiers throughout its development. It is most commonly referred to by its research code, MLN4924, and was also designated TAK-924 by Takeda Pharmaceuticals.[18] Its formally assigned generic name is Pevonedistat.

The compound's precise chemical structure is defined by its International Union of Pure and Applied Chemistry (IUPAC) name: amino]pyrrolo[2,3-d]pyrimidin-7-yl]-2-hydroxycyclopentyl]methyl sulfamate.[13] This nomenclature reflects a complex molecule with specific stereochemistry that is critical for its biological activity. For computational and database cross-referencing, its structure is also represented by standardized identifiers such as the Simplified Molecular Input Line Entry System (SMILES) and the International Chemical Identifier (InChI) and its corresponding hashed key (InChI Key).[18]

To ensure unambiguous identification across scientific and regulatory databases, Pevonedistat is cataloged under several key reference numbers. These include CAS Number: 905579-51-3; DrugBank Accession Number: DB11759; PubChem Compound ID (CID): 16720766; FDA Unique Ingredient Identifier (UNII): S3AZD8D215; and ChEMBL ID: CHEMBL1231160.[13]

2.2 Physicochemical Characteristics

Pevonedistat's molecular composition is described by the chemical formula C21​H25​N5​O4​S.[18] This corresponds to a

molecular weight (or molar mass) consistently reported as approximately 443.5 g/mol or 443.52 Da.[15]

In its purified form, Pevonedistat is typically supplied as a lyophilized white powder or solid.[15] Its solubility characteristics are crucial for its formulation and administration. The compound is soluble in organic solvents such as dimethyl sulfoxide (DMSO), with reported solubilities of 10 mg/mL to 20 mg/mL, and in ethanol.[15] However, its solubility in aqueous solutions is limited, which necessitates its formulation as a concentrate for solution for intravenous infusion for clinical use.[13]

From a chemical classification perspective, Pevonedistat is a multifaceted molecule. Its core structure is a pyrrolopyrimidine, and it is further characterized as a secondary amino compound, a member of cyclopentanols, a sulfamidate, and a derivative of indane.[13] This complex structure contributes to its specific binding affinity and inhibitory activity against its target enzyme.

For ease of reference, the key identifiers and physicochemical properties of Pevonedistat are consolidated in Table 1.

Table 1: Key Identifiers and Physicochemical Properties of Pevonedistat

PropertyValueSource Snippet(s)
Generic NamePevonedistat13
Synonyms/CodesMLN4924, MLN-4924, TAK-92418
DrugBank IDDB1175913
CAS Number905579-51-313
PubChem CID1672076618
IUPAC Nameamino]pyrrolo[2,3-d]pyrimidin-7-yl]-2-hydroxycyclopentyl]methyl sulfamate13
Chemical FormulaC21​H25​N5​O4​S18
Molecular Weight~443.5 g/mol18
Chemical ClassPyrrolopyrimidine, Indane, Sulfamidate13
SolubilitySoluble in DMSO (e.g., 10-20 mg/mL)15
InChI KeyMPUQHZXIXSTTDU-QXGSTGNESA-N18

3.0 Pharmacology and Detailed Mechanism of Action

3.1 The NEDD8-Activating Enzyme (NAE) as a Therapeutic Target

The therapeutic activity of Pevonedistat is rooted in its inhibition of the neddylation pathway, a crucial post-translational modification system that parallels the well-known ubiquitination pathway.[15] The central process involves the covalent attachment of the ubiquitin-like protein NEDD8 (Neural precursor cell expressed developmentally down-regulated 8) to target proteins, thereby altering their function, localization, or stability.[14]

The entire cascade is initiated by a single E1-type enzyme, the NEDD8-Activating Enzyme (NAE).[11] NAE is a heterodimer composed of a regulatory subunit, APPBP1, and a catalytic subunit, UBA3.[15] In an ATP-dependent reaction, NAE activates NEDD8 and prepares it for transfer to an E2-conjugating enzyme (UBC12) and subsequently to a substrate via an E3 ligase.[18]

The most critical and well-characterized substrates of neddylation are the cullin proteins.[24] Cullins are molecular scaffolds that form the core of

Cullin-RING E3 ubiquitin ligases (CRLs), the largest family of E3 ligases, which are responsible for targeting approximately 20% of all proteasome-degraded proteins.[11] The covalent attachment of NEDD8 to a conserved lysine residue on the cullin subunit is an absolute requirement for CRL activation.[16] This modification induces a conformational change that positions the substrate for efficient ubiquitination and subsequent degradation by the proteasome.[24] By controlling the activity of hundreds of CRLs, the neddylation pathway governs the turnover of a vast array of proteins involved in fundamental cellular processes, including cell cycle progression, DNA replication, signal transduction, and stress responses.[11] Its central role in maintaining protein homeostasis makes NAE an attractive target for anticancer therapy.

3.2 Molecular Interaction and NAE Inhibition

Pevonedistat exerts its effect through a highly specific and potent molecular interaction with NAE. Structurally, Pevonedistat is an analog of adenosine 5'-monophosphate (AMP), which is a natural intermediate in the NAE-catalyzed reaction.[15] Pevonedistat binds to the nucleotide-binding site within the catalytic pocket of the UBA3 subunit of NAE.[18]

The enzyme mistakenly utilizes Pevonedistat in place of AMP to react with NEDD8. This reaction forms a covalent and functionally irreversible NEDD8-pevonedistat adduct that remains tightly bound to the enzyme.[16] Unlike the transient, high-energy NEDD8-AMP intermediate that is poised for the next step in the cascade, the stable NEDD8-pevonedistat adduct is a dead-end product. It effectively traps the enzyme in an inactive state, preventing the transfer of activated NEDD8 to the E2 enzyme and thereby halting the entire neddylation cascade at its initial step.[16]

This inhibition is both potent and highly selective. Biochemical assays have determined the half-maximal inhibitory concentration (IC50​) of Pevonedistat against NAE to be 4.7 nM.[13] Its selectivity is remarkable when compared to its activity against the homologous E1 enzymes of related pathways. The

IC50​ values for the ubiquitin-activating enzyme (UAE) and the SUMO-activating enzyme (SAE) are 1.5 µM and 8.2 µM, respectively.[22] This represents a selectivity of over 300-fold for NAE over UAE and over 1700-fold for NAE over SAE, minimizing off-target effects on these other critical cellular pathways.

3.3 Downstream Cellular Consequences

The direct pharmacological effect of NAE inhibition—the blockade of the neddylation cascade—triggers a cascade of downstream cellular events that collectively contribute to Pevonedistat's antitumor activity.

The primary consequence is the global inactivation of CRLs, as their cullin scaffolds can no longer be neddylated.[16] This functional shutdown leads to the rapid

accumulation of numerous CRL substrate proteins that would otherwise be ubiquitinated and degraded.[11] The accumulation of these specific proteins, many of which are key regulators of cellular proliferation and survival, induces a state of multifaceted proteotoxic stress. This multi-pronged attack on cancer cell biology explains the drug's potent preclinical activity. The process unfolds through several interconnected mechanisms:

  1. Cell Cycle Disruption and DNA Damage: A key CRL substrate is CDT-1, a crucial DNA replication licensing factor whose levels must be tightly controlled to prevent genomic instability.[11] NAE inhibition leads to CDT-1 accumulation, which causes aberrant re-firing of DNA replication origins within a single S-phase. This "DNA re-replication" is a catastrophic event that leads to extensive DNA damage, activation of the DNA damage response (e.g., CHK1 activation), and ultimately, cell cycle arrest, typically in the G2 or M phase.[11]
  2. Impairment of DNA Repair: Beyond inducing damage, Pevonedistat also compromises the cell's ability to repair it. Neddylation is known to be required for the proper function of at least two major DNA repair pathways: nucleotide excision repair (NER) and non-homologous end joining (NHEJ).[18] By inhibiting NAE, Pevonedistat creates an induced deficiency in these repair mechanisms. This dual action—inducing DNA damage while simultaneously blocking its repair—can be particularly lethal to cancer cells, which often harbor pre-existing defects in other DNA repair pathways, creating a scenario of synthetic lethality.[18] This provides a strong mechanistic rationale for combining Pevonedistat with conventional DNA-damaging chemotherapies like cytarabine, as Pevonedistat can potentiate their effects by preventing the repair of drug-induced DNA lesions.[11]
  3. Inhibition of Pro-Survival Signaling: NAE inhibition also disrupts critical pro-survival signaling pathways. For example, the CRL complex SCFβ−TrCP is responsible for degrading phospho-IκBα, the inhibitor of the NF-κB transcription factor.[11] By preventing phospho-IκBα degradation, Pevonedistat effectively traps NF-κB in the cytoplasm, suppressing the activity of this key pathway that promotes cancer cell survival, proliferation, and inflammation.[16]
  4. Induction of Apoptosis: The culmination of overwhelming DNA damage, cell cycle catastrophe, and suppression of survival signals is the induction of apoptosis, or programmed cell death.[13] The cellular response to Pevonedistat, however, may be context-dependent. Studies in neuroblastoma cell lines have shown that the ultimate cytotoxic mechanism is dependent on the status of the tumor suppressor p53.[26] Cells with wild-type p53 underwent G0-G1 cell-cycle arrest followed by robust apoptosis. In contrast, cells with mutant or deficient p53 underwent G2-M arrest and rereplication without immediate apoptosis.[26] This finding suggests that the genetic background of a tumor could dictate its sensitivity and mode of death in response to NAE inhibition. This mechanistic heterogeneity may have significant clinical implications, as it suggests that patient responses in a genetically diverse population, such as those in the PANTHER trial, could vary substantially. The lack of biomarker-driven patient selection based on factors like p53 status may have contributed to the dilution of the treatment effect observed in the broad trial population.

4.0 Pharmacokinetic Profile: Absorption, Distribution, Metabolism, and Excretion (ADME)

4.1 Route of Administration and Disposition

The clinical formulation of Pevonedistat is designed for intravenous (IV) infusion, typically administered over a 60-minute period.[27] This route of administration ensures complete (100%) bioavailability and circumvents potential issues related to oral absorption and first-pass metabolism.[30]

Pharmacokinetic (PK) analyses from multiple Phase 1 studies have consistently shown that plasma concentrations of Pevonedistat decline in a bi-exponential manner following the end of the infusion.[27] The drug exhibits linear pharmacokinetics, with exposure (as measured by the area under the plasma concentration-time curve, AUC) increasing proportionately with the dose across the clinically evaluated range.[27] The disposition of Pevonedistat is characterized by a relatively short mean terminal elimination half-life (

t1/2​) in plasma, estimated to be between 5 and 8.4 hours.[27] This kinetic profile results in little to no drug accumulation with the intermittent dosing schedules used in clinical trials (e.g., on Days 1, 3, and 5 of a 21- or 28-day cycle), as the drug is substantially cleared from the plasma between doses.[27]

4.2 Distribution, Metabolism, and Excretion Pathways

The movement and fate of Pevonedistat in the body have been characterized through dedicated clinical studies, including a human mass balance study using radiolabeled [14C]-pevonedistat (NCT03057366).[28]

Distribution: A defining and unusual characteristic of Pevonedistat's pharmacokinetics is its extensive and preferential distribution into whole blood. The mean whole-blood-to-plasma concentration ratio for total drug exposure (AUC) was determined to be 40.8, indicating that the vast majority of the drug in circulation is not free in the plasma but is sequestered within blood components, primarily red blood cells.[28] This partitioning is thought to be mediated by binding to carbonic anhydrase II, an enzyme highly abundant within erythrocytes.[28] This phenomenon suggests that red blood cells may act as a circulating reservoir for Pevonedistat. This reservoir could buffer high peak plasma concentrations immediately after infusion and then slowly release the drug back into circulation, potentially providing a more sustained exposure at target tissues, such as the bone marrow, than would be predicted by its short plasma half-life alone. This sustained local exposure may help explain the clinical activity observed with an intermittent dosing schedule.

Metabolism: Pevonedistat undergoes extensive metabolism, which is the primary driver of its clearance.[28] Analysis of radiolabeled drug showed that unchanged Pevonedistat accounted for only 41-44% of the total drug-related material in circulation, with the remainder composed of various metabolites.[28] The low renal clearance of the parent drug points to hepatic metabolism as the main elimination pathway. While initial in vitro studies implicated the cytochrome P450 3A (CYP3A) family of enzymes in its metabolism, subsequent clinical drug-drug interaction studies demonstrated that co-administration with strong or moderate CYP3A inhibitors did not lead to clinically meaningful increases in Pevonedistat exposure.[28] This suggests that multiple, potentially redundant, metabolic pathways are involved in its biotransformation.

Excretion: The human mass balance study provided a definitive account of the drug's excretion routes. Over a one-week period following a single IV dose of [14C]-pevonedistat, a mean of 94% of the total administered radioactivity was recovered.[28] The excretion was well-balanced between two primary routes:

53% of the dose was recovered in the feces (indicative of biliary excretion of the drug and/or its metabolites) and 41% was recovered in the urine.[28] Importantly, the renal clearance of the unchanged parent drug was found to be minimal, accounting for only about 2.5% of its total plasma clearance.[28] This confirms that while metabolites are cleared renally, the direct excretion of active Pevonedistat via the kidneys is a very minor pathway.

5.0 Clinical Development and Efficacy Evaluation

5.1 Early Phase Development and Safety Profile

Pevonedistat's clinical journey began with a series of Phase 1 trials designed to assess its safety, determine the appropriate dose, and gather preliminary evidence of antitumor activity. These initial studies evaluated Pevonedistat both as a single agent and in combination with standard-of-care chemotherapies across a range of advanced malignancies, including solid tumors, lymphoma, and multiple myeloma.[13]

A pivotal Phase 1b dose-escalation study (NCT01814826) focused on the target population of older (≥60 years), treatment-naïve patients with AML who were unfit for intensive chemotherapy.[12] This study established the combination of Pevonedistat with azacitidine as a promising and tolerable regimen. The

Recommended Phase 2 Dose (RP2D) of Pevonedistat was determined to be 20 mg/m² administered as an IV infusion on Days 1, 3, and 5 of a 28-day cycle, in combination with the standard dose of azacitidine.[12] The primary dose-limiting toxicities (DLTs) were transient, reversible elevations in liver transaminases (AST/ALT), and the combination did not appear to exacerbate the myelosuppressive effects of azacitidine.[12] The clinical activity in this study was highly encouraging, with an intent-to-treat (ITT)

overall response rate (ORR) of 50%, including a composite complete remission (CR/CRi) rate of 42%.[12]

To support a global development program, a dedicated Phase 1/1b study (NCT02782468) was conducted in East Asian patients with AML or MDS.[17] This trial confirmed that the pharmacokinetic and safety profiles of Pevonedistat, both alone and with azacitidine, were comparable between East Asian and Western patient populations.[17] It independently established the identical RP2D of 20 mg/m² for Pevonedistat with azacitidine, providing the necessary evidence to include patients from this region in subsequent global trials.[17] Consistent with other early studies, objective responses were observed only in the combination arm (a 45% response rate in AML patients), whereas single-agent Pevonedistat resulted in stable disease at best, solidifying the rationale for combination therapy moving forward.[17]

5.2 The Promise of Phase 2 (NCT02610777): A Turning Point

The randomized, open-label, proof-of-concept Phase 2 study (NCT02610777) was the critical inflection point in Pevonedistat's development, generating the compelling data that propelled it into Phase 3 testing.[37] The study randomized 120 patients with HR-MDS, HR-CMML, or low-blast AML to receive either Pevonedistat plus azacitidine (P+A) or azacitidine alone (A).[38]

While the study was underpowered for its primary endpoint of Overall Survival (OS) in the ITT population, and the difference did not reach statistical significance (median OS 21.8 months for P+A vs. 19.0 months for A; HR 0.80), the results within the prespecified HR-MDS subgroup (n=67) were striking and clinically meaningful.[5] In this key patient population, the addition of Pevonedistat to azacitidine demonstrated substantial improvements across multiple endpoints:

  • Event-Free Survival (EFS): The combination led to a statistically significant and clinically relevant improvement in EFS, defined as the time to transformation to AML or death. The median EFS was 20.2 months for the P+A arm compared to 14.8 months for the azacitidine alone arm (Hazard Ratio 0.54; 95% CI 0.29–1.00; p=0.045).[5]
  • Overall Survival (OS): A strong trend toward improved OS was observed, with a median OS of 23.9 months with P+A versus 19.1 months with azacitidine alone (HR 0.70; 95% CI 0.39–1.27; p=0.240).[38]
  • Complete Remission (CR) Rate: The rate of complete remission was nearly doubled in the combination arm, at 52% versus 27% for azacitidine monotherapy.[39]
  • Duration of Response (DoR): For patients who responded, the durability of that response was substantially longer with the combination, with a median DoR of 34.6 months compared to just 13.1 months with azacitidine alone.[39]

Crucially, these efficacy benefits were achieved with a safety profile that was comparable to azacitidine monotherapy, with no increase in myelosuppression or other significant toxicities.[5] The strength and consistency of these data in the HR-MDS subgroup were the basis for the U.S. FDA's decision to grant Pevonedistat a

Breakthrough Therapy Designation, accelerating its path toward a pivotal trial.[2]

5.3 The Pivotal PANTHER Trial (NCT03268954): Analysis of Phase 3 Failure

Based on the highly promising Phase 2 results, the global, randomized, open-label Phase 3 PANTHER trial was initiated to provide definitive evidence of Pevonedistat's efficacy.[42] The study enrolled 454 patients with newly diagnosed HR-MDS, HR-CMML, or low-blast AML, randomizing them to P+A versus azacitidine alone.[1]

In a major setback for the program, Takeda announced in September 2021 that the PANTHER trial failed to meet its primary endpoint of improving EFS in the ITT population.[44] The final analysis showed a median EFS of 17.7 months for the P+A arm versus 15.7 months for the azacitidine arm, a difference that was not statistically significant (HR 0.968; 95% CI 0.757–1.238; p=0.557).[1]

Most critically, the robust signal of benefit seen in the HR-MDS subgroup in the Phase 2 trial was not replicated. In the larger HR-MDS cohort of the PANTHER trial (n=324), the addition of Pevonedistat did not lead to a statistically significant improvement in either EFS or OS. The median EFS was 19.2 months with P+A versus 15.6 months with azacitidine alone (HR 0.887; p=0.431), and the median OS was 21.6 months versus 17.5 months, respectively (HR 0.785; p=0.092).[1] Furthermore, the impressive doubling of the CR rate seen in Phase 2 vanished; in Phase 3, the CR rate was numerically lower in the combination arm (24%) than in the control arm (32%).[1] The safety profile remained consistent with prior studies, with no new concerns arising.[1] The stark contrast between the Phase 2 promise and the Phase 3 reality is illustrated in Table 2.

Table 2: Comparative Efficacy Endpoints in Phase 2 vs. Phase 3 Trials for the HR-MDS Subgroup

Efficacy EndpointPhase 2 (NCT02610777)Phase 3 (PANTHER, NCT03268954)Source Snippet(s)
Median Event-Free Survival (EFS)20.2 months (P+A) vs. 14.8 months (A)19.2 months (P+A) vs. 15.6 months (A)1
Hazard Ratio (HR) for EFS0.54 (p=0.045)0.887 (p=0.431)1
Median Overall Survival (OS)23.9 months (P+A) vs. 19.1 months (A)21.6 months (P+A) vs. 17.5 months (A)1
Hazard Ratio (HR) for OS0.70 (p=0.240)0.785 (p=0.092)1
Complete Remission (CR) Rate52% (P+A) vs. 27% (A)24% (P+A) vs. 32% (A)1

5.4 Reconciling Disparate Outcomes: A Critical Analysis

The failure of Pevonedistat to transition from a successful Phase 2 to a positive Phase 3 trial is not attributable to a simple lack of biological activity but rather to a complex interplay of factors related to trial design, evolving standards of care, and the inherent heterogeneity of the disease.

A primary contributor to the negative outcome was the unexpectedly high performance of the azacitidine-alone control arm in the PANTHER trial. As shown in Table 2, the CR rate for azacitidine monotherapy jumped from 27% in Phase 2 to 32% in Phase 3, and the median EFS and OS also appeared robust. This "overperformance" raised the statistical bar for the combination to demonstrate a significant incremental benefit. Clinical trial investigators have suggested that the PANTHER study protocol included stricter language discouraging premature dose reductions or treatment delays for azacitidine. This likely ensured that patients in the control arm received a more optimal and consistent standard-of-care regimen than in the previous Phase 2 study or in historical cohorts, thereby improving their outcomes and narrowing the therapeutic gap for Pevonedistat to overcome.[47]

Perhaps the most illuminating finding from the PANTHER trial came from a post-hoc analysis that examined outcomes based on duration of therapy. This analysis revealed a statistically significant overall survival benefit for the P+A combination in HR-MDS patients who were able to remain on treatment for more than three, and particularly for more than six, cycles. For patients receiving more than six cycles, the median OS was 27.1 months with the combination versus 22.5 months with azacitidine alone (p=0.008).[1] This finding strongly suggests that the clinical benefit of NAE inhibition is not immediate but requires sustained therapeutic pressure over multiple treatment cycles to manifest in deep, durable responses. The overall result of the trial was likely diluted by a substantial proportion of patients who discontinued treatment early due to adverse events, disease progression, or other reasons, and therefore did not receive therapy long enough to derive this long-term benefit. The smaller Phase 2 study may have, by chance or design, enrolled a patient population more capable of tolerating prolonged therapy, leading to the initial strong positive signal. The larger, more heterogeneous global Phase 3 trial revealed the real-world challenge: the benefit is conditional on treatment persistence, a factor that must be addressed in the design of future studies.

5.5 Exploration of Novel Combination Strategies

Following the PANTHER trial results, the clinical development strategy for Pevonedistat shifted toward exploring its potential in different combinations and disease settings. A strong preclinical rationale emerged for combining Pevonedistat with the BCL-2 inhibitor venetoclax.[48] Upregulation of the anti-apoptotic protein MCL-1 is a known mechanism of resistance to venetoclax. Pevonedistat, through its complex downstream effects, can neutralize MCL-1, suggesting a synergistic cytotoxic effect when combined with venetoclax.[49]

This hypothesis was tested in clinical trials. A Phase 1 study of the triplet combination of Pevonedistat, azacitidine, and venetoclax in patients with relapsed/refractory AML (NCT04172844) found the regimen to be safe and demonstrated encouraging preliminary activity. The ORR was 46.7% for the overall cohort, rising to an impressive 71.4% CR rate in the subset of patients who had not previously been treated with an HMA/venetoclax combination.[50]

However, this early promise was tempered by the results of a randomized Phase 2 study (NCT04266795) that evaluated the same triplet combination against the standard-of-care doublet of azacitidine plus venetoclax in newly diagnosed, unfit AML patients.[33] This study was stopped prematurely after the negative PANTHER trial results were announced. The final analysis of the available data showed no significant difference in the primary endpoint of EFS or in OS between the triplet and doublet arms.[33] While an exploratory analysis hinted at higher response rates in a small subgroup of patients with IDH mutations, the overall result did not support the addition of Pevonedistat to the azacitidine/venetoclax backbone in the frontline AML setting.[33]

6.0 Regulatory Trajectory and Current Status

6.1 United States FDA Journey

The regulatory pathway for Pevonedistat in the United States has been marked by early promise followed by a halt in progress. Recognizing the high unmet need in acute myeloid leukemia, the U.S. Food and Drug Administration (FDA) granted Pevonedistat Orphan Drug Designation for the treatment of AML on February 4, 2011.[51] This designation is intended to provide incentives, such as tax credits and market exclusivity, to encourage the development of drugs for rare diseases affecting fewer than 200,000 people in the U.S.

The most significant regulatory milestone was achieved on July 30, 2020, when the FDA granted Pevonedistat Breakthrough Therapy Designation for the treatment of patients with HR-MDS.[2] This designation is designed to expedite the development and review of drugs that are intended to treat a serious condition and for which preliminary clinical evidence indicates that the drug may demonstrate substantial improvement over available therapy on a clinically significant endpoint.[6] The decision was based entirely on the compelling final results from the Phase 2 Pevonedistat-2001 study (NCT02610777), particularly the significant improvement in EFS and the doubling of the CR rate in the HR-MDS subgroup.[5]

Despite these encouraging designations, Pevonedistat remains an investigational agent and is not FDA approved for any indication.[51] The failure of the pivotal Phase 3 PANTHER trial to confirm the Phase 2 results effectively nullified the data package that would have supported a New Drug Application (NDA) for the HR-MDS indication.

6.2 European Medicines Agency (EMA) History

Pevonedistat also pursued a regulatory path in Europe through the European Medicines Agency (EMA). The EMA granted Orphan Designation for Pevonedistat for the treatment of myelodysplastic syndromes on December 14, 2018 (designation number EU/3/18/2120).[13] This was followed by a second

Orphan Designation for the treatment of acute myeloid leukaemia on July 25, 2019 (EU/3/19/2186).[54] These designations acknowledged the life-threatening nature of the diseases and the potential for Pevonedistat to provide a significant benefit to affected patients.

However, in a definitive regulatory and strategic development, the sponsor, Takeda, submitted a request to the EMA for the withdrawal of both orphan designations. This withdrawal became effective in February 2022.[53] This action is a direct and telling consequence of the negative PANTHER trial results announced in September 2021.[44] The timing, approximately five months after the trial failure, is not coincidental. Maintaining an orphan designation involves ongoing administrative and financial commitments. A company re-evaluates a drug's development program after a pivotal trial fails. The decision to actively withdraw the designations, rather than simply letting them lapse or not filing for marketing authorization, represents a clear strategic signal that the company no longer foresees a viable path to approval in Europe with the existing data. This move effectively dismantles the established regulatory framework for Pevonedistat in the EU and indicates a fundamental re-strategizing of the drug's global development program.

7.0 Synthesis and Future Outlook

7.1 Critical Assessment of Pevonedistat's Therapeutic Potential

Pevonedistat stands as a compound with a strong scientific rationale and a complex clinical history. Its primary strength lies in its novel, first-in-class mechanism of action. As a highly potent and selective inhibitor of NAE, it targets a fundamental cellular process—neddylation—that is critical for the survival and proliferation of cancer cells.[10] Its ability to induce multifaceted proteotoxic stress through concurrent cell cycle disruption, DNA damage induction, and inhibition of pro-survival signaling provides a compelling basis for its antitumor activity. Furthermore, its clinical safety profile has been consistently shown to be manageable, allowing for its combination with the standard-of-care agent azacitidine without introducing significant additive myelosuppression or overlapping toxicities.[10]

The definitive weakness of Pevonedistat, however, is the unequivocal failure of its pivotal Phase 3 PANTHER trial to demonstrate a statistically significant clinical benefit over azacitidine alone.[10] This outcome has cast significant doubt on its efficacy as a broad treatment for an unselected population of patients with HR-MDS and related myeloid neoplasms. The inability to replicate the striking results of the Phase 2 study highlights the inherent risks of drug development and the challenges of translating promising mid-stage signals into definitive late-stage success.

7.2 Future Research Directions

While the path to approval for Pevonedistat in HR-MDS based on the PANTHER trial data is closed, the scientific rationale for targeting the neddylation pathway remains valid. The future of Pevonedistat, if one exists, will depend on a more nuanced and targeted development strategy focused on two key areas:

  1. Biomarker-Driven Patient Selection: The heterogeneity of clinical responses, coupled with preclinical data showing context-dependent mechanisms of cell death (e.g., p53 status), strongly suggests that a "one-size-fits-all" approach is suboptimal.[26] Future clinical trials must be designed to prospectively identify patient subsets most likely to benefit from NAE inhibition. This will require the development and validation of predictive biomarkers. Potential candidates could include the mutational status of key genes like TP53, baseline expression levels of critical CRL substrates (e.g., CDT-1), or gene expression signatures associated with DNA damage repair pathways or cell cycle regulation. Identifying a responsive subpopulation is likely the only viable path forward for this agent.
  2. Rational Combination Strategies: The exploration of novel combinations must continue, but with a more rigorous, mechanistically-driven approach. The initial attempt to combine Pevonedistat with azacitidine and venetoclax in frontline AML did not succeed, but this does not preclude its potential utility in other settings.[33] There may be a role for such triplet combinations in the relapsed/refractory setting, where overcoming resistance is paramount, or in specific, molecularly defined subgroups (such as the IDH-mutated AML population where a small signal was observed).[33] Further preclinical work is essential to identify the most synergistic therapeutic partners and the optimal clinical context for their evaluation.

7.3 Concluding Remarks

The development trajectory of Pevonedistat offers a sobering but invaluable lesson in modern oncology drug development. It exemplifies the perilous "valley of death" between promising Phase 2 data and the rigorous demands of a confirmatory Phase 3 trial. The story underscores that a trial's failure is not always due to a lack of drug activity but can result from a complex confluence of factors, including the continual improvement in the standard-of-care control arm, the inherent biological and genetic heterogeneity of the patient population, and a potential mismatch between the drug's mechanism and the trial's primary endpoint timeline.

The post-hoc finding that sustained treatment duration was linked to improved survival is a critical takeaway, suggesting that for some novel agents, the therapeutic benefit may only emerge over time. This highlights the need for future trial designs to incorporate strategies that manage toxicity effectively to maximize treatment persistence. While the future of Pevonedistat as a broad-spectrum therapy for HR-MDS is now highly uncertain, the neddylation pathway remains a compelling and scientifically validated target for cancer therapy. The extensive clinical and biological data generated from the Pevonedistat program will undoubtedly serve as a crucial foundation, informing the development of next-generation neddylation inhibitors and guiding the design of more sophisticated, biomarker-driven clinical trials in these challenging hematologic malignancies.

Works cited

  1. Pevonedistat plus azacitidine vs azacitidine alone in higher-risk MDS/chronic myelomonocytic leukemia or low-blast-percentage AML, accessed August 19, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9631625/
  2. Takeda Announces U.S. FDA Breakthrough Therapy Designation Granted for Pevonedistat for the Treatment of Patients with Higher-Risk Myelodysplastic Syndromes (HR-MDS), accessed August 19, 2025, https://www.takedaoncology.com/news/news-releases/takeda-announces-u.s.-fda-breakthrough-therapy-designation-granted-for-pevonedistat-for-the-treatment-of-patients-with-higher-risk-myelodysplastic-syndromes-hr-mds/
  3. Living with Higher-Risk Myelodysplastic Syndromes - Conquer: the journey informed, accessed August 19, 2025, https://conquer-magazine.com/issues/special-issues/december-2020-living-with-higher-risk-myelodysplastic-syndromes
  4. Current Standard of Care in Patients with Myelodysplastic Syndromes and Future Perspectives, accessed August 19, 2025, https://onco-hema.healthbooktimes.org/article/25627-current-standard-of-care-in-patients-with-myelodysplastic-syndromes-and-future-perspectives
  5. FDA Grants Breakthrough Therapy Designation to Pevonedistat in Higher-Risk MDS, accessed August 19, 2025, https://www.targetedonc.com/view/fda-grants-breakthrough-therapy-designation-to-pevonedistat-in-higher-risk-mds
  6. FDA Grants Breakthrough Therapy Designation for Pevonedistat to Treat Higher-Risk MDS, accessed August 19, 2025, https://www.cancernetwork.com/view/fda-grants-breakthrough-therapy-designation-for-pevonedistat-to-treat-higher-risk-mds
  7. Treatment of high-risk myelodysplastic syndromes - PMC, accessed August 19, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11788630/
  8. Myelodysplastic Syndromes Treatment (PDQ®)–Patient Version - National Cancer Institute, accessed August 19, 2025, https://www.cancer.gov/types/myeloproliferative/patient/myelodysplastic-treatment-pdq
  9. Therapeutic options in high-risk myelodysplastic syndrome | Maksymowicz | Hematology in Clinical Practice - Via Medica Journals, accessed August 19, 2025, https://journals.viamedica.pl/hematology_in_clinical_practice/article/view/HCP.a2022.0017
  10. The development of pevonedistat in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML): hope or hype? - PMC, accessed August 19, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9310330/
  11. The NEDD8-Activating Enzyme Inhibitor MLN4924 Disrupts ..., accessed August 19, 2025, https://aacrjournals.org/clincancerres/article/21/2/439/245070/The-NEDD8-Activating-Enzyme-Inhibitor-MLN4924
  12. Pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor, combined with azacitidine in patients with AML | Blood | American Society of Hematology, accessed August 19, 2025, https://ashpublications.org/blood/article/131/13/1415/36624/Pevonedistat-a-first-in-class-NEDD8-activating
  13. Pevonedistat | C21H25N5O4S | CID 16720766 - PubChem, accessed August 19, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Pevonedistat
  14. What is Pevonedistat used for? - Patsnap Synapse, accessed August 19, 2025, https://synapse.patsnap.com/article/what-is-pevonedistat-used-for
  15. MLN-4924, NEDD8-activating enzyme inhibitor (CAS 905579-51-3) - Abcam, accessed August 19, 2025, https://www.abcam.com/en-us/products/biochemicals/mln-4924-nedd8-activating-enzyme-inhibitor-ab216470
  16. Pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor, sensitizes cancer cells to VSVΔ51 oncolytic virotherapy - PubMed Central, accessed August 19, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10638453/
  17. Pevonedistat in East Asian patients with acute myeloid leukemia or ..., accessed August 19, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9097234/
  18. Pevonedistat - Wikipedia, accessed August 19, 2025, https://en.wikipedia.org/wiki/Pevonedistat
  19. Pevonedistat: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed August 19, 2025, https://go.drugbank.com/drugs/DB11759
  20. MLN 4924 | Other Ligases - Tocris Bioscience, accessed August 19, 2025, https://www.tocris.com/products/mln-4924_6499
  21. PEVONEDISTAT (PD003365, MPUQHZXIXSTTDU-QXGSTGNESA-N) - Probes & Drugs, accessed August 19, 2025, https://www.probes-drugs.org/compound/PD003365/
  22. MLN4924 (Pevonedistat, CAS Number: 905579-51-3) | Cayman Chemical, accessed August 19, 2025, https://www.caymanchem.com/product/15217/mln4924
  23. MLN4924 (Pevonedistat) #85923 - Cell Signaling Technology, accessed August 19, 2025, https://www.cellsignal.com/products/activators-inhibitors/mln4924-pevonedistat/85923
  24. Pevonedistat, a NEDD8-activating enzyme inhibitor, is active in ..., accessed August 19, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4778163/
  25. The NEDD8-activating enzyme inhibitor MLN4924 reduces ischemic brain injury in mice | PNAS, accessed August 19, 2025, https://www.pnas.org/doi/10.1073/pnas.2111896119
  26. The Anti-Tumor Activity of the NEDD8 Inhibitor Pevonedistat in Neuroblastoma, accessed August 19, 2025, https://escholarship.org/uc/item/6698v0xs
  27. Population pharmacokinetics of pevonedistat alone or in combination with standard of care in patients with solid tumours or haematological malignancies - PubMed Central, accessed August 19, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6848912/
  28. Phase I study assessing the mass balance, pharmacokinetics, and ..., accessed August 19, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7960626/
  29. A Study of [14 C]-Pevonedistat in Participants With Advanced Solid Tumors, accessed August 19, 2025, https://clinicaltrials.gov/study/NCT03057366
  30. Pharmacokinetics: Definition & Use in Drug Development - Allucent, accessed August 19, 2025, https://www.allucent.com/resources/blog/what-pharmacokinetics-and-adme
  31. Definition of carbon C 14 pevonedistat - NCI Drug Dictionary, accessed August 19, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/carbon-c-14-pevonedistat
  32. Malignant Lymphomas Completed Phase 1 Trials for Pevonedistat (DB11759) - DrugBank, accessed August 19, 2025, https://go.drugbank.com/indications/DBCOND0046362/clinical_trials/DB11759?phase=1&status=completed
  33. Full article: Azacitidine and venetoclax with or without pevonedistat in patients with newly diagnosed acute myeloid leukemia - Taylor & Francis Online, accessed August 19, 2025, https://www.tandfonline.com/doi/full/10.1080/10428194.2024.2431878
  34. A Study of Pevonedistat in Adult East Asian Participants - ClinicalTrials.gov, accessed August 19, 2025, https://clinicaltrials.gov/study/NCT02782468
  35. (PDF) Pevonedistat in East Asian patients with acute myeloid leukemia or myelodysplastic syndromes: a phase 1/1b study to evaluate safety, pharmacokinetics and activity as a single agent and in combination with azacitidine - ResearchGate, accessed August 19, 2025, https://www.researchgate.net/publication/360533326_Pevonedistat_in_East_Asian_patients_with_acute_myeloid_leukemia_or_myelodysplastic_syndromes_a_phase_11b_study_to_evaluate_safety_pharmacokinetics_and_activity_as_a_single_agent_and_in_combination_wit
  36. Pevonedistat in East Asian patients with acute myeloid leukemia or myelodysplastic syndromes: a phase 1/1b study to evaluate safety, pharmacokinetics and activity as a single agent and in combination with azacitidine - PubMed, accessed August 19, 2025, https://pubmed.ncbi.nlm.nih.gov/35545778/
  37. An Efficacy and Safety Study of Pevonedistat Plus Azacitidine Versus Single-Agent Azacitidine in Participants With Higher-Risk Myelodysplastic Syndromes (HR MDS), Chronic Myelomonocytic Leukemia (CMML) and Low-Blast Acute Myelogenous Leukemia (AML) | ClinicalTrials.gov, accessed August 19, 2025, https://clinicaltrials.gov/study/NCT02610777
  38. Phase II study of pevonedistat (P) + azacitidine (A) versus A in patients (pts) with higher-risk myelodysplastic syndromes (MDS)/chronic myelomonocytic leukemia (CMML), or low-blast acute myelogenous leukemia (LB AML) (NCT02610777). - ASCO, accessed August 19, 2025, https://www.asco.org/abstracts-presentations/ABSTRACT298543
  39. Takeda Announces Compelling Data from the Phase 2 Trial of Pevonedistat Plus Azacitidine in Patients with Higher-Risk MDS, accessed August 19, 2025, https://www.takeda.com/newsroom/newsreleases/2020/takeda-announces-compelling-data-from-the-phase-2-trial-of-pevonedistat-plus-azacitidine-in-patients-with-higher-risk-mds/
  40. PHASE 2 STUDY OF PEVONEDISTAT + AZACITIDINE VERSUS AZACITIDINE IN PATIENTS WITH HIGHER-RISK MYELODYSPLASTIC SYNDROMES/CHRONIC MYELOMONOCYTIC LEUKEMIA OR LOW-BLAST ACUTE MYELOGENOUS LEUKEMIA (NCT02610777) | Hematology, Transfusion and Cell Therapy, accessed August 19, 2025, http://www.htct.com.br/en-phase-2-study-of-pevonedistat-articulo-S2531137920307616
  41. Pevonedistat plus Azacitidine Improves Outcomes versus Azacitidine Monotherapy in High-Risk MDS, accessed August 19, 2025, https://jhoponline.com/meeting-highlights/pevonedistat-plus-azacitidine-improves-outcomes-versus-azacitidine-monotherapy-in-high-risk-mds
  42. Pevonedistat Active Not Recruiting Phase 3 Trials for Acute Myeloid Leukemia Treatment, accessed August 19, 2025, https://go.drugbank.com/drugs/DB11759/clinical_trials?conditions=DBCOND0029883&phase=3&purpose=treatment&status=active_not_recruiting
  43. Pevonedistat Plus Azacitidine Versus Single-Agent Azacitidine as First-Line Treatment for Participants With Higher-Risk Myelodysplastic Syndromes (HR MDS), Chronic Myelomonocytic Leukemia (CMML), or Low-Blast Acute Myelogenous Leukemia (AML) | ClinicalTrials.gov, accessed August 19, 2025, https://clinicaltrials.gov/study/NCT03268954
  44. Takeda's pevonedistat fails to meet primary goal in Phase III leukaemia trial, accessed August 19, 2025, https://www.clinicaltrialsarena.com/news/takeda-pevonedistat-leukaemia-trial/
  45. Takeda Provides Update on Phase 3 PANTHER (Pevonedistat-3001) Trial, accessed August 19, 2025, https://www.takeda.com/newsroom/newsreleases/2021/takeda-provides-update-on-phase-3-panther-pevonedistat-3001-trial/
  46. Pevonedistat plus azacitidine vs azacitidine alone in higher-risk MDS/chronic myelomonocytic leukemia or low-blast-percentage AML | Blood Advances | American Society of Hematology, accessed August 19, 2025, https://ashpublications.org/bloodadvances/article-abstract/6/17/5132/485649
  47. PANTHER: No Significant Benefit for Pevonedistat Plus Azacitidine in Higher-Risk Myelodysplastic Syndrome - The ASCO Post, accessed August 19, 2025, https://ascopost.com/issues/february-25-2022/no-significant-benefit-for-pevonedistat-plus-azacitidine-in-higher-risk-myelodysplastic-syndrome/
  48. Pevonedistat, venetoclax and azacitidine for older AML patients - YouTube, accessed August 19, 2025, https://www.youtube.com/watch?v=-hhFQazLBPc
  49. A Randomized Phase 2 Study of Pevonedistat, Venetoclax, and Azacitidine Versus Venetoclax Plus Azacitidine in Adults with Newly Diagnosed Acute Myeloid Leukemia (AML) Who Are Unfit for Intensive Chemotherapy - American Society of Hematology, accessed August 19, 2025, https://ashpublications.org/blood/article/136/Supplement%201/34/470801/A-Randomized-Phase-2-Study-of-Pevonedistat
  50. A phase I study of pevonedistat, azacitidine, and venetoclax in patients with relapsed/refractory acute myeloid leukemia | Haematologica, accessed August 19, 2025, https://haematologica.org/article/view/haematol.2024.285014
  51. Search Orphan Drug Designations and Approvals - FDA, accessed August 19, 2025, https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=331510
  52. Oncology (Cancer)/Hematologic Malignancies Approval Notifications - FDA, accessed August 19, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/oncology-cancerhematologic-malignancies-approval-notifications
  53. EU/3/18/2120 - orphan designation for treatment of myelodysplastic ..., accessed August 19, 2025, https://www.ema.europa.eu/en/medicines/human/orphan-designations/eu-3-18-2120
  54. EU/3/19/2186 - orphan designation for treatment of acute myeloid ..., accessed August 19, 2025, https://www.ema.europa.eu/en/medicines/human/orphan-designations/eu-3-19-2186
  55. A therapeutic drug for myelodysplastic syndrome: pevonedistat, accessed August 19, 2025, http://www.lcywzlzz.com/EN/10.3969/j.issn.1672-3384.2024.03.004

Published at: August 19, 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.