MedPath

Evofosfamide Advanced Drug Monograph

Published:Sep 13, 2025

Generic Name

Evofosfamide

Drug Type

Small Molecule

Chemical Formula

C9H16Br2N5O4P

CAS Number

918633-87-1

Evofosfamide (TH-302): A Comprehensive Monograph on a Hypoxia-Activated Prodrug from Pivotal Trial Failure to Immunotherapeutic Revival

1.0 Introduction to Evofosfamide: A Hypoxia-Activated Prodrug

1.1 The Challenge of Tumor Hypoxia in Oncology

A fundamental characteristic of solid tumors is their tendency to outgrow their vascular supply, leading to the formation of regions with low oxygen concentration, a state known as hypoxia.[1] This hypoxic tumor microenvironment (TME) is not merely a passive consequence of tumor growth but an active driver of malignant progression and therapeutic resistance. Hypoxic cancer cells undergo significant metabolic reprogramming, often driven by the stabilization of the transcription factor Hypoxia-Inducible Factor 1α (HIF-1α), which promotes a shift towards anaerobic glycolysis for energy production.[3] Clinically, tumor hypoxia is strongly correlated with increased tumor aggressiveness, a higher propensity for metastasis, and profound resistance to a wide range of standard-of-care cancer treatments.[5] Both conventional chemotherapy and, particularly, radiation therapy often depend on the presence of molecular oxygen to generate the cytotoxic reactive oxygen species that mediate their efficacy. Consequently, the oxygen-deprived cells within the tumor core are frequently spared from treatment, serving as a reservoir for disease recurrence and progression.[5] Overcoming the barrier of tumor hypoxia remains one of the most significant and persistent challenges in modern oncology.

1.2 Rationale and Development of Evofosfamide (TH-302)

Instead of attempting to overcome or reverse hypoxia, a more elegant strategy involves exploiting this unique feature of the TME to achieve tumor-selective drug activation. This is the principle behind Hypoxia-Activated Prodrugs (HAPs), a class of therapeutics designed to remain largely inert in well-oxygenated, healthy tissues but to undergo bioreductive activation to release a potent cytotoxin specifically within the hypoxic regions of a tumor.[1]

Evofosfamide, also known as TH-302, was developed as a second-generation HAP based on this principle. It is a molecular conjugate of two key components: a 2-nitroimidazole moiety that acts as a hypoxia-sensing "trigger," and a masked, potent DNA alkylating agent, bromo-isophosphoramide mustard (Br-IPM), which serves as the cytotoxic "warhead".[1] The development of Evofosfamide by Threshold Pharmaceuticals Inc. was the result of systematic structure-activity relationship (SAR) studies aimed at improving upon existing phosphoramidate mustard chemotherapies like ifosfamide. A critical optimization was the replacement of the chlorine atoms in the mustard effector with bromine atoms. This chemical modification was found to increase the cytotoxic potency of the released alkylating agent by approximately tenfold, without compromising its high degree of hypoxic selectivity.[1] This rational design positioned Evofosfamide as a highly promising agent with the potential for targeted tumor destruction while minimizing the systemic toxicity associated with conventional chemotherapy.

1.3 Overview of Developmental Trajectory and Current Status

The developmental history of Evofosfamide is a compelling narrative of scientific promise, clinical disappointment, and strategic reinvention. Its elegant mechanism and strong preclinical data attracted significant industry interest, culminating in a major global co-development agreement between Threshold Pharmaceuticals and Merck KGaA in 2012.[1] The drug advanced rapidly into late-stage clinical trials for multiple solid tumors.

However, in 2015, the program suffered a major setback when two pivotal Phase III trials—one in advanced pancreatic cancer and another in soft tissue sarcoma—both failed to meet their primary endpoint of improving overall survival.[11] These failures led to the termination of the collaboration and the discontinuation of the drug's development for these indications.

Despite these results, the unique biology of Evofosfamide continued to attract interest. In 2020, ImmunoGenesis, Inc. acquired the rights to the compound, initiating a strategic pivot based on a new therapeutic hypothesis.[15] The focus shifted from viewing Evofosfamide solely as a direct cytotoxic agent to repositioning it as a "hypoxia-reversal agent" and a modulator of the tumor microenvironment. The current strategy is to use Evofosfamide to eliminate hypoxic, immunosuppressive cells, thereby remodeling the TME to make immunologically "cold" tumors more susceptible to the effects of immune checkpoint inhibitors. Evofosfamide remains an investigational drug and is not approved for any indication, but it is currently being evaluated in clinical trials under this new immuno-oncology paradigm.[15]

2.0 Physicochemical Properties and Chemical Synthesis

2.1 Chemical Structure and Identifiers

Evofosfamide is a small molecule drug classified as a member of the imidazoles, a phosphorodiamidate ester, and an organobromine compound.[17] Its unambiguous identification is established through a variety of chemical and regulatory identifiers, which are consolidated in Table 1.

Table 1: Chemical and Physical Identifiers of Evofosfamide

Identifier TypeValueSource(s)
NameEvofosfamide1
SynonymsTH-302, TH302, HAP 3022
TypeSmall Molecule18
DrugBank IDDB060911
CAS Number918633-87-11
IUPAC Name2-bromo-N-[(2-bromoethylamino)-[(3-methyl-2-nitroimidazol-4-yl)methoxy]phosphoryl]ethanamine1
Molecular FormulaC9​H16​Br2​N5​O4​P17
Molecular Weight449.04 g/mol17
InChI KeyUGJWRPJDTDGERK-UHFFFAOYSA-N1
SMILESCN1C(=CN=C1N+[O-])COP(=O)(NCCBr)NCCBr
Other IDsPubChem CID: 11984561, UNII: 8A9RZ3HN8W, KEGG: D10704, ChEMBL: CHEMBL260046

2.2 Physical and Chemical Properties

Evofosfamide presents as an off-white to pale yellow solid with a melting point of 97-98°C. Its molecular formula is

C9​H16​Br2​N5​O4​P, corresponding to a molecular weight of 449.04 g/mol and a monoisotopic mass of 446.93067 Da. The elemental analysis is C, 24.07%; H, 3.59%; Br, 35.59%; N, 15.60%; O, 14.25%; P, 6.90%.

Solubility studies indicate that Evofosfamide is soluble in organic solvents such as dimethylformamide (DMF), dimethyl sulfoxide (DMSO), and ethanol at concentrations of 30 mg/mL. Its solubility in aqueous solutions is limited, with a reported solubility of 0.3 mg/mL in phosphate-buffered saline (PBS) at pH 7.2. The compound is stable enough for shipment at ambient temperature.

Computed physicochemical properties are consistent with those of a small molecule drug candidate. It has 10 rotatable bonds and a topological polar surface area of 111.32 to 121.13 Ų. With 2 hydrogen bond donors and 9 Lipinski hydrogen bond acceptors, it does not violate Lipinski's Rule of Five, suggesting favorable properties for oral bioavailability, although it has been developed for intravenous administration.

2.3 Key Aspects of Synthesis and Structure-Activity Relationships (SAR)

The synthesis of Evofosfamide is a targeted chemical process involving two principal steps. The first is the preparation of the 2-nitroimidazole moiety, which serves as the key bioreductive group responsible for the drug's hypoxia-selective activation. The second step involves covalently linking this trigger to a brominated derivative of isophosphoramide mustard, the cytotoxic effector.

The final chemical structure of Evofosfamide was the result of deliberate SAR studies designed to optimize the potency and selectivity of an earlier generation of HAPs. Researchers began with the chemical scaffold of known and effective phosphoramidate mustards, such as ifosfamide, which contains chloroethyl groups. Initial work produced a chlorinated version of the HAP (TH-281), which demonstrated a high hypoxia cytotoxicity ratio (HCR). Subsequent SAR studies revealed that substituting the two chlorine atoms on the mustard effector with bromine atoms resulted in a significant, approximately 10-fold increase in cytotoxic potency. This enhancement was achieved while preserving the high degree of hypoxic selectivity, leading to the selection of Evofosfamide (TH-302) as the lead clinical candidate. Further research has explored other structural modifications, including changes to substituents on the nitrogen atoms and the position of the nitro group, with some novel derivatives demonstrating even greater antitumor activity than Evofosfamide in vitro against ovarian cancer and glioblastoma cell lines.

3.0 Mechanism of Action: Selective Targeting of the Hypoxic Tumor Microenvironment

3.1 Bioreductive Activation Pathway

Evofosfamide functions as a prodrug that is largely biologically inert under the normoxic conditions found in healthy tissues. Its activation is contingent upon the unique, low-oxygen biochemical environment characteristic of solid tumors. The activation cascade is initiated by a one-electron reduction of the 2-nitroimidazole ring of the Evofosfamide molecule. This reaction is catalyzed by a range of ubiquitous intracellular one-electron-donating reductases, with NADPH:cytochrome P450 oxidoreductase (POR) being a key enzyme implicated in this process. The transfer of a single electron to the nitro group generates a highly reactive and unstable radical anion intermediate of the parent drug. The fate of this radical anion is the critical determinant of the drug's activity and is dictated entirely by the local concentration of molecular oxygen.

3.2 The Role of the 2-Nitroimidazole Trigger

The 2-nitroimidazole component of Evofosfamide serves as a sophisticated oxygen sensor. Under normoxic conditions (typically defined as oxygen levels >0.5%), the radical anion intermediate is extremely short-lived. It rapidly reacts with abundant molecular oxygen in a process that transfers the electron to oxygen, generating a superoxide radical and regenerating the original, inactive Evofosfamide prodrug. This futile process, known as redox cycling, effectively prevents the accumulation of the radical anion and the subsequent release of the cytotoxic agent, thereby protecting healthy, well-oxygenated tissues from damage.

In stark contrast, under the severe hypoxic conditions prevalent in many solid tumors (<0.5% O2), the scarcity of molecular oxygen prevents this rapid re-oxidation. The radical anion intermediate persists for a sufficient duration to undergo an irreversible, spontaneous fragmentation. This oxygen-dependent differential stability is the cornerstone of Evofosfamide's tumor selectivity. The drug's efficacy is therefore critically dependent on a specific therapeutic window of hypoxia—oxygen levels must be low enough to permit activation, but the cells must remain metabolically active enough to provide the necessary reductase enzymes. This complex interplay between oxygen tension and cellular metabolic state within the heterogeneous tumor microenvironment likely contributes to the variable clinical responses observed.

3.3 Generation and Action of Bromo-Isophosphoramide Mustard (Br-IPM)

The irreversible fragmentation of the Evofosfamide radical anion under hypoxia results in the cleavage of the molecule, releasing two products: the active cytotoxic effector, bromo-isophosphoramide mustard (Br-IPM), and an inactive azole derivative. Br-IPM is a powerful, bifunctional DNA alkylating agent, belonging to the nitrogen mustard class of compounds. As a bifunctional agent, it possesses two reactive bromoethyl groups. These groups can form highly electrophilic aziridinium ion intermediates that covalently bind to nucleophilic sites on DNA, with a preference for the N7 position of guanine bases. Because each Br-IPM molecule can react twice, it is capable of forming both intrastrand crosslinks (linking two bases on the same DNA strand) and, more critically, interstrand crosslinks (linking bases on opposite DNA strands).

3.4 Downstream Cellular Consequences: DNA Damage, Cell Cycle Arrest, and Apoptosis

The formation of interstrand DNA crosslinks is one of the most cytotoxic forms of DNA damage, as it physically prevents the separation of the DNA double helix. This blockage effectively halts essential cellular processes, including DNA replication and transcription, leading to catastrophic genomic instability. The cell's DNA damage response (DDR) pathways are immediately activated, leading to the phosphorylation of the histone variant H2AX to form γH2AX, a well-established biomarker of DNA double-strand breaks and severe DNA damage.

In an attempt to repair this damage, the cell cycle is arrested, primarily at the G0/G1 checkpoint, to prevent the propagation of damaged DNA to daughter cells. This arrest is mediated by the downregulation of key cell cycle proteins, including cyclins D1/2/3 and cyclin-dependent kinases CDK4/6. If the DNA damage induced by Br-IPM is too extensive for the cell's repair machinery to handle, the apoptotic cell death program is initiated. This is characterized by the activation of the caspase cascade, including initiator caspases (caspase-8, -9) and executioner caspases (caspase-3), and the subsequent cleavage of key cellular substrates like poly (ADP-ribose) polymerase (PARP).

Preclinical studies have revealed that the primary mechanism for repairing the damage caused by Evofosfamide is the homology-dependent repair (HDR) pathway. Consequently, cell lines with deficiencies in key HDR proteins, such as BRCA1, BRCA2, or FANCA, exhibit markedly increased sensitivity to Evofosfamide under hypoxic conditions. This dependency on a specific DNA repair pathway suggests a strong scientific rationale for combining Evofosfamide with inhibitors of parallel repair pathways, such as PARP inhibitors, to induce synthetic lethality and enhance its antitumor effect.

3.5 The "Bystander Effect": Mechanisms and Controversies

A key component of the therapeutic rationale for Evofosfamide was the "bystander effect," a phenomenon wherein the active Br-IPM, once released in a hypoxic cell, could diffuse into the surrounding tumor tissue and kill adjacent, better-oxygenated cancer cells that would otherwise be unaffected. This concept was supported by three-dimensional tumor spheroid and multicellular layer models, which suggested that Evofosfamide could exert a significant effect beyond the zone of activation. This proposed mechanism was attractive because it implied that the drug could eliminate not only the hypoxic core but also the normoxic, proliferative rim of a tumor, potentially leading to more comprehensive tumor control.

However, the significance of this bystander effect has been challenged by more recent pharmacological studies. These investigations have suggested that the active metabolites, Br-IPM and its downstream product IPM, are hydrophilic molecules that are unable to efficiently pass across cell membranes. This would severely limit their ability to diffuse out of the cell of origin and kill neighboring cells. An alternative explanation for the observed cell killing in better-oxygenated regions is that at high concentrations of the parent prodrug, a low level of residual activation and Br-IPM formation can still occur even in the presence of oxygen. The true contribution of the bystander effect to the overall in vivo antitumor activity of Evofosfamide therefore remains an area of scientific debate.

4.0 Preclinical Pharmacology and Efficacy

The clinical development of Evofosfamide was underpinned by a robust and extensive body of preclinical evidence demonstrating its potent, hypoxia-selective antitumor activity across a wide array of cancer models.

Table 2: Summary of Key Preclinical Studies

Cancer ModelStudy TypeCombination AgentKey FindingSource(s)
H460 NSCLC CellsIn VitroNoneHypoxia Cytotoxicity Ratio (HCR) of ~270; IC50 of 0.019 µM (hypoxia) vs. 5.1 µM (normoxia)
32 Human Cancer Cell LinesIn VitroNoneEnhanced cytotoxicity under hypoxia consistently observed across all cell lines
Multiple Xenografts (NSCLC, SCLC, Melanoma, Prostate, Pancreatic)In VivoNone>40% tumor growth inhibition at 50 mg/kg
H460 NSCLC XenograftIn VivoNoneUp to 89% tumor growth inhibition at 50 mg/kg
MIA PaCa-2 Pancreatic Orthotopic XenograftIn VivoGemcitabine96% inhibition of primary tumor growth; significantly extended survival
SCCVII & HT29 XenograftsIn VivoIonizing RadiationSignificant benefit observed in both tumor models
Neuroblastoma Metastatic ModelIn VivoNoneSignificantly increased mouse survival compared to vehicle
Canine Glioma Murine ModelIn VivoNone (vs. Temozolomide)Significantly reduced tumor development compared to controls and temozolomide

4.1 In Vitro Assessment of Hypoxic Selectivity and Potency

In vitro studies consistently demonstrated the remarkable hypoxia-selective cytotoxicity of Evofosfamide. Across a panel of 32 different human cancer cell lines, the compound was significantly more potent under hypoxic conditions (N2​ or <0.5% O2​) than under normoxic (21% O2​) conditions. This selectivity is quantified by the Hypoxia Cytotoxicity Ratio (HCR), the ratio of the IC50 value in normoxia to the IC50 value in hypoxia. For Evofosfamide, this ratio was exceptionally high. For example, in H460 non-small cell lung cancer (NSCLC) cells, the IC50 was 5.1 µM in normoxia but only 0.019 µM in hypoxia, yielding an HCR of approximately 270. Similar high levels of selectivity were observed in numerous other cell lines, confirming the fidelity of its hypoxia-activated mechanism at the cellular level.

4.2 In Vivo Efficacy in Monotherapy Xenograft Models

The potent and selective activity observed in vitro translated effectively to in vivo preclinical models. When administered as a monotherapy, Evofosfamide produced significant dose-dependent tumor growth inhibition in a wide range of human cancer xenografts. In models of Calu-6 NSCLC, H82 small cell lung cancer, A375 melanoma, PC-3 prostate cancer, and BxPC-3 pancreatic cancer, a dose of 50 mg/kg inhibited tumor growth by over 40%. In the H460 NSCLC xenograft model, the same dose regimen led to tumor growth suppression of up to 89%. Efficacy was also demonstrated in models of neuroendocrine prostate cancer (84.5% tumor growth inhibition) and aggressive neuroblastoma, where it significantly increased survival in a metastatic model. These studies provided strong proof-of-concept for its potential as a single-agent therapy targeting hypoxic tumors.

4.3 Synergistic Effects in Combination with Chemotherapy and Radiotherapy

Given that Evofosfamide targets the hypoxic, therapy-resistant cell populations that limit the efficacy of conventional treatments, it was a logical candidate for combination therapy. Preclinical studies demonstrated strong synergistic or additive effects when Evofosfamide was combined with standard-of-care agents. The combination with gemcitabine was particularly effective in a MIA PaCa-2 human pancreatic cancer orthotopic xenograft model, resulting in a 96% inhibition of primary tumor growth and a significant extension of survival time compared to either agent alone. Similarly, when combined with ionizing radiation, Evofosfamide showed significant therapeutic benefit in both SCCVII and HT29 tumor models, validating the concept of using it to eliminate radioresistant hypoxic cells.

4.4 Impact on the Tumor Microenvironment and Metabolic Pathways

The therapeutic effects of Evofosfamide extend beyond its direct cytotoxicity. By selectively eliminating the highly metabolic and oxygen-consuming cells within hypoxic regions, the drug can profoundly remodel the tumor microenvironment. A key consequence of this is tumor reoxygenation. Studies using quantitative pO2 imaging by electron paramagnetic resonance (EPR) demonstrated that Evofosfamide treatment led to a global improvement in oxygenation in responsive pancreatic cancer xenografts. This effect was shown to be a result of decreased oxygen demand from the tumor, rather than an improvement in blood perfusion or vascularity.

Furthermore, by targeting HIF-1α-positive cells, Evofosfamide can disrupt the aberrant metabolic state of tumors. In canine glioma models, Evofosfamide treatment inhibited glycolytic ATP production, leading to a decrease in total cellular ATP and a suppression of tumor development. The drug has also been shown to restore type I interferon signaling that is suppressed by hypoxia in breast cancer cells, thereby enhancing their susceptibility to being killed by natural killer (NK) cells. This ability to remodel the TME—by alleviating hypoxia, normalizing metabolism, and reversing immune suppression—was likely an underappreciated aspect of its mechanism during its initial development but now forms the central thesis for its current evaluation in immuno-oncology.

5.0 Clinical Development and Efficacy Analysis

5.1 Overview of the Clinical Trial Program

The promising preclinical data for Evofosfamide propelled it into a broad and ambitious clinical development program. It was investigated as a monotherapy and in combination with standard-of-care agents across a diverse range of malignancies, including pancreatic cancer, soft tissue sarcoma, multiple myeloma, leukemia, melanoma, and head and neck cancer. The program progressed rapidly through Phase I and II studies, ultimately leading to two pivotal, international Phase III trials.

Table 3: Overview of Major Clinical Trials for Evofosfamide

Trial ID / NCT #PhaseIndicationTreatment ArmsPrimary EndpointOutcome SummarySource(s)
MAESTRO / NCT01746979IIIPancreatic Ductal AdenocarcinomaEvofosfamide + Gemcitabine vs. Placebo + GemcitabineOverall SurvivalFailed to meet primary endpoint (p=0.059), but showed significant improvement in PFS and ORR
TH-CR-406/SARC021 / NCT01440088IIISoft Tissue SarcomaEvofosfamide + Doxorubicin vs. Doxorubicin aloneOverall SurvivalFailed to meet primary endpoint (p=0.527); no significant improvement in PFS
NCT01149915IAdvanced Leukemia (AML/ALL)Evofosfamide MonotherapyMTD / SafetyLimited activity (6% ORR) in heavily pretreated patients
NCT00495144IAdvanced MelanomaEvofosfamide MonotherapySafety / EfficacyShowed signs of activity (19% PR, 33% SD)
NCT03098160IAdvanced Solid TumorsEvofosfamide + IpilimumabSafety / EfficacyEvidence of therapeutic activity (16.7% PR, 66.7% SD)
NCT06782555I/IICRPC, Pancreatic, HNSCCEvofosfamide + Balstilimab + ZalifrelimabSafety / EfficacyRecruiting; first patient dosed March 2025

5.2 Phase III MAESTRO Trial in Pancreatic Cancer: Design and Outcomes

The MAESTRO study (NCT01746979) was a large-scale, international, randomized, double-blind, placebo-controlled Phase III trial designed to evaluate the efficacy of Evofosfamide in combination with gemcitabine for the first-line treatment of patients with locally advanced unresectable or metastatic pancreatic ductal adenocarcinoma (PDAC). A total of 693 patients were randomized to receive either Evofosfamide (340 mg/m²) plus gemcitabine (1,000 mg/m²) or a matching placebo plus gemcitabine, administered intravenously on days 1, 8, and 15 of a 28-day cycle.

The primary endpoint of the study was overall survival (OS). The trial narrowly missed this endpoint, showing a modest but not statistically significant improvement in the Evofosfamide arm. The median OS was 8.7 months for the Evo/Gem group compared to 7.6 months for the Placebo/Gem group (Hazard Ratio = 0.84; 95% CI: 0.71–1.01; p = 0.059).

Despite failing to meet the primary endpoint, the study did demonstrate statistically significant improvements in key secondary endpoints. Median progression-free survival (PFS) was significantly longer in the Evofosfamide arm at 5.5 months versus 3.7 months in the placebo arm (HR = 0.77; 95% CI: 0.65–0.92; p = 0.004). The confirmed objective response rate (ORR) was also significantly higher with Evofosfamide, at 15% compared to 9% (Odds Ratio = 1.90; 95% CI: 1.16–3.12; p = 0.009).

5.3 Phase III TH-CR-406/SARC021 Trial in Soft Tissue Sarcoma: Design and Outcomes

The TH-CR-406/SARC021 study (NCT01440088) was an international, randomized, open-label Phase III trial that enrolled 640 patients with locally advanced, unresectable, or metastatic soft-tissue sarcoma (STS) who had not received prior chemotherapy. Patients were randomized to receive either the standard-of-care agent doxorubicin (75 mg/m²) alone or doxorubicin in combination with Evofosfamide (300 mg/m²).

The primary endpoint was overall survival. The trial failed to demonstrate any benefit for the addition of Evofosfamide. The median OS was slightly shorter in the combination arm at 18.4 months compared to 19.0 months in the doxorubicin-alone arm (HR = 1.06; 95% CI: 0.88–1.29; p = 0.527).

Secondary endpoints also showed little to no benefit. There was no statistically significant improvement in median PFS (6.3 months for the combination vs. 6.0 months for doxorubicin alone; HR = 0.85; p = 0.099). The only positive signal was a statistically significant improvement in ORR, which was 28% in the combination arm versus 18% in the doxorubicin arm (p = 0.0026).

5.4 Analysis of Phase I/II Trials in Other Malignancies

Beyond the pivotal trials, Evofosfamide was evaluated in several other cancer types with varying results:

  • Melanoma: A Phase I monotherapy study (NCT00495144) in 36 patients with advanced melanoma showed encouraging signs of activity, with 7 patients (19%) achieving a partial response and 12 patients (33%) achieving stable disease.
  • Leukemia: A Phase I study (NCT01149915) in 49 heavily pretreated patients with relapsed/refractory acute myeloid leukemia (AML) or acute lymphoblastic leukemia (ALL) demonstrated that the drug was tolerable but had limited single-agent activity. The combined overall response rate was only 6%, though the drug did reduce bone marrow hypoxia markers.
  • Head and Neck Squamous Cell Cancer (HNSCC): A Phase I trial (NCT03098160) was conducted to evaluate Evofosfamide in combination with the immunotherapy agent ipilimumab in patients with HPV-negative HNSCC, among other solid tumors. In 18 evaluable patients across all tumor types, the combination resulted in a 16.7% partial response rate and a 66.7% stable disease rate, providing early evidence of therapeutic activity for this combination.

5.5 Critical Analysis of Pivotal Trial Failures

The failure of Evofosfamide to meet its primary OS endpoint in two large, well-conducted Phase III trials was a profound disappointment and led to the cessation of its joint development by Threshold and Merck KGaA. The inability to convert improvements in secondary endpoints like PFS and ORR (as seen in the MAESTRO trial) into a statistically significant overall survival benefit is a common challenge in oncology drug development and is often insufficient for regulatory approval. While the SARC021 trial in STS showed a clear lack of efficacy, the near-miss in the MAESTRO trial for pancreatic cancer prompted deeper investigation. This post-hoc analysis uncovered a critical flaw in the trial's execution related to the drug's pharmacokinetics, suggesting that the failure may have been due to operational factors rather than a fundamental lack of biological activity, a topic explored in detail in Section 7.0.

6.0 Safety and Tolerability Profile

6.1 Summary of Adverse Events from Clinical Trials

The safety profile of Evofosfamide has been characterized across numerous clinical trials. The most frequently reported and clinically significant adverse events (AEs) were hematologic and mucosal toxicities, consistent with the effects of a DNA alkylating agent.

In the Phase III MAESTRO trial, hematologic AEs such as neutropenia, thrombocytopenia, and anemia were more frequent in the arm receiving Evofosfamide plus gemcitabine compared to the placebo plus gemcitabine arm. However, common non-hematologic AEs like nausea (47%), decreased appetite (35%), and vomiting (33%) occurred at similar rates in both arms. Importantly, AEs leading to death were slightly lower in the Evofosfamide arm (9%) compared to the placebo arm (11%).

The Phase III SARC021 trial provided a clearer picture of the added toxicity of Evofosfamide, as it was an open-label comparison against doxorubicin alone. The combination arm exhibited significantly higher rates of several Grade ≥3 AEs.

Table 5: Summary of Grade ≥3 Adverse Events in Pivotal Phase III Trials

Adverse EventMAESTRO: Evo + Gem (%)MAESTRO: Placebo + Gem (%)SARC021: Evo + Dox (%)SARC021: Dox Alone (%)Source(s)
AnemiaMore FrequentBaseline4821
NeutropeniaMore FrequentBaseline1530
Febrile NeutropeniaNot SpecifiedNot Specified1811
ThrombocytopeniaMore FrequentBaseline141
StomatitisNot SpecifiedNot Specified82
Serious AEsNot SpecifiedNot Specified4632

6.2 Dose-Limiting Toxicities and Maximum Tolerated Dose

Phase I dose-escalation studies established the dose-limiting toxicities (DLTs) and maximum tolerated dose (MTD) for Evofosfamide. In a study in patients with advanced leukemia (NCT01149915), the DLTs were primarily mucosal. For a daily 30-60 minute infusion schedule, Grade 3 esophagitis was the DLT at 550 mg/m², establishing an MTD of 460 mg/m². For a continuous 120-hour infusion, Grade 3 stomatitis and hyperbilirubinemia were the DLTs at 460 mg/m², with an MTD of 330 mg/m². In a Phase I monotherapy study in advanced melanoma, skin and mucosal toxicities were also identified as dose-limiting.

6.3 Comparative Safety Insights

An important aspect of Evofosfamide's profile is its potential for a more favorable safety margin compared to related, non-targeted alkylating agents. A preclinical study directly compared Evofosfamide with ifosfamide, a structurally related prodrug that requires hepatic activation. The study found that at doses producing an equal level of antitumor efficacy (measured by body weight loss), ifosfamide caused significantly more severe hematologic toxicity. Conversely, at doses matched for an equal level of hematologic toxicity, Evofosfamide demonstrated superior antitumor activity. This finding supports the core rationale of HAPs: by restricting activation to the hypoxic tumor, the systemic exposure of healthy tissues, particularly the bone marrow, to the active cytotoxin is reduced, potentially widening the therapeutic window.

7.0 Pharmacokinetic Profile and Formulation Considerations

7.1 Preclinical Pharmacokinetic Data

Preclinical studies in animal models provided initial characterization of Evofosfamide's pharmacokinetic (PK) properties. These studies indicated that the drug has a short plasma half-life of approximately 12.3 minutes and a high clearance rate of 2.29 L/h/kg. There was also evidence of significant biliary excretion and/or secretion into the gut, suggesting multiple routes of elimination.

7.2 Human Pharmacokinetics from Phase I and II Studies

Phase I studies in humans established the initial safety, tolerability, and general PK characteristics of Evofosfamide. The subsequent randomized Phase II trial in pancreatic cancer (part of the lead-up to the MAESTRO study) was particularly informative. It evaluated two doses of Evofosfamide (240 mg/m² and 340 mg/m²) in combination with gemcitabine. The results showed a clear dose-response relationship, with the 340 mg/m² dose demonstrating a significant improvement in both ORR and PFS compared to the 240 mg/m² dose and to gemcitabine alone. This established 340 mg/m² as the optimal dose to carry forward into the pivotal Phase III trial.

7.3 The Impact of Formulation Change on Pharmacokinetics in the MAESTRO Trial

A critical and likely decisive factor in the failure of the Phase III MAESTRO trial was an unexpected pharmacokinetic issue stemming from a change in the drug's formulation. Between the completion of the successful Phase II study and the start of the Phase III study, a new ethanol-based formulation was introduced to improve the drug product's solubility and manufacturing characteristics. While seemingly a routine process improvement, a post-hoc analysis of PK data from the trials revealed that this change had a dramatic and detrimental effect on drug exposure in patients.

The analysis showed that the systemic exposure achieved with the 340 mg/m² dose using the new formulation in Phase III was substantially lower than the exposure achieved with the same dose in Phase II. In fact, the PK profile (both maximum concentration, Cmax​, and area under the curve, AUC) of the 340 mg/m² dose in Phase III was nearly identical to that of the less effective 240 mg/m² dose from Phase II.

Table 4: Comparative Pharmacokinetic Parameters from Phase II vs. Phase III MAESTRO Studies

Trial / DoseN (patients)Geometric Mean Cmax​ (µg/mL)Geometric Mean AUC (µg-h/mL)Source(s)
Phase II / 240 mg/m² + Gem47 (Cmax), 44 (AUC)5.325.33
Phase II / 340 mg/m² + Gem51 (Cmax), 47 (AUC)9.278.94
Phase III / 340 mg/m² + Gem317 (Cmax), 302 (AUC)6.346.02

This stark discrepancy provides a compelling explanation for the trial's outcome. The patients in the pivotal Phase III study were effectively underdosed relative to the exposure level that had demonstrated superior efficacy in Phase II. The efficacy results from the Phase III trial (median OS of 8.7 months, median PFS of 5.5 months) closely mirrored the results from the suboptimal 240 mg/m² dose arm in Phase II (median OS of 8.7 months, median PFS of 5.6 months). This suggests that the MAESTRO trial did not fail because the drug's biological mechanism was ineffective, but rather because a critical pharmaceutical development step—ensuring bioequivalence after a formulation change—was overlooked or inadequately performed, leading to a failure to deliver a therapeutic dose in the pivotal study. This represents a significant learning in drug development, underscoring that pharmaceutical science is as critical to success as the underlying biology.

8.0 Strategic Repositioning and Future Outlook

8.1 The Rationale for Hypoxia Reversal in Immuno-Oncology

Following the setbacks in Phase III, the development of Evofosfamide has been revitalized under a new strategic framework focused on immuno-oncology (I-O). This repositioning is grounded in the evolving understanding of the TME. It is now well-established that tumor hypoxia is a primary driver of an intensely immunosuppressive microenvironment. Hypoxia promotes the recruitment of myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs), while physically and functionally excluding cytotoxic T-cells, the primary effectors of cancer immunotherapy. This creates an "immune-excluded" or "cold" tumor phenotype that is intrinsically resistant to immune checkpoint inhibitors (ICIs) like anti-PD-1 and anti-CTLA-4 antibodies.

The new therapeutic hypothesis posits that Evofosfamide can function as a potent TME modulator. By selectively killing hypoxic cells, it can alleviate the overall oxygen demand within the tumor, leading to reoxygenation and a reversal of the hypoxic state. This remodeling of the TME is theorized to dismantle the immunosuppressive barrier, allowing for the infiltration and activation of T-cells, thereby converting "cold" tumors into "hot," immune-responsive ones, and synergizing with ICI therapy.

8.2 Current Clinical Trials with Immune Checkpoint Inhibitors

This new strategy is being actively pursued in the clinic. In December 2020, ImmunoGenesis, Inc. acquired the rights to Evofosfamide to develop it specifically as a hypoxia-reversal agent for I-O combinations.

An initial proof-of-concept came from a Phase I study (NCT03098160) combining Evofosfamide with the anti-CTLA-4 antibody ipilimumab. In patients with advanced solid tumors, including prostate cancer and HNSCC, the combination was found to be tolerable and showed clear evidence of therapeutic activity, with an objective response rate of 16.7% and a disease stabilization rate of 66.7% in a heavily pretreated population.

Building on this, a new Phase 1/2 trial (NCT06782555) was initiated in March 2025. This study is evaluating a triplet combination of Evofosfamide with two modern ICIs: balstilimab (an anti-PD-1 antibody) and zalifrelimab (an anti-CTLA-4 antibody). The trial is enrolling patients with immunologically "cold" tumors known for their resistance to ICIs, including metastatic castration-resistant prostate cancer (CRPC), pancreatic cancer, and HPV-negative HNSCC. The study follows a dose-escalation design to determine the optimal dose of Evofosfamide for this combination, followed by a dose-expansion phase to assess safety, PK, and antitumor activity. The estimated completion date for this pivotal study is January 2028.

8.3 Potential as a Modulator of the Tumor Immune Microenvironment

The scientific basis for this new approach is supported by both preclinical and clinical evidence. Preclinical models have shown that targeting hypoxia with Evofosfamide can improve T-cell infiltration and lead to tumor regression when combined with ICIs. Crucially, translational data from the Phase I trial with ipilimumab provided the first clinical validation of this mechanism. Analysis of patient samples revealed that those who responded to the combination therapy showed improved peripheral T-cell proliferation and, most importantly, increased infiltration of T-cells directly into the hypoxic regions of their tumors. This provides direct evidence that Evofosfamide can, in humans, help overcome the physical and functional barriers that hypoxia erects against an effective anti-tumor immune response.

8.4 Future Research Directions and Unanswered Questions

The ongoing clinical program will be critical in answering key questions that will determine the future of Evofosfamide. The primary question is whether the triplet combination can generate durable clinical responses in "cold" tumor types where ICI monotherapy has largely failed. A second crucial area of investigation will be the identification of predictive biomarkers. Data from the ipilimumab study suggested that pre-existing immune gene signatures could predict response, while hypermetabolic tumors predicted progression, highlighting the need for sophisticated patient selection strategies. Finally, careful attention must be paid to the drug's dose, schedule, and formulation to ensure that the pharmacokinetic lessons from the MAESTRO trial are applied and that patients receive an optimal biological dose for TME modulation. The success of this new chapter for Evofosfamide will depend on demonstrating that its unique mechanism can finally be translated into meaningful clinical benefit for patients with these difficult-to-treat cancers.

9.0 Comprehensive Summary and Expert Insights

9.1 Synthesis of Evofosfamide's Profile: A Drug of Two Narratives

The trajectory of Evofosfamide is best understood as a story of two distinct, yet interconnected, narratives. The first narrative is that of a brilliantly designed, second-generation hypoxia-activated prodrug. It was built on a strong scientific rationale, optimized through meticulous SAR studies, and supported by a wealth of compelling preclinical data. It represented a direct and elegant approach to targeting a core driver of malignancy and therapy resistance. This narrative culminated in large-scale Phase III trials, where it ultimately failed to deliver on its promise, a failure now understood to be heavily confounded by a critical, and likely avoidable, error in pharmaceutical development related to a formulation change.

The second narrative is one of scientific and strategic revival. It recasts Evofosfamide not merely as a targeted cytotoxin, but as a first-in-class modulator of the tumor microenvironment. This new identity is built upon a deeper understanding of the interplay between tumor hypoxia and immune suppression—a field of biology that was in its infancy when the drug was first developed. By leveraging the drug's proven ability to eliminate hypoxic cells and reoxygenate the TME, the new strategy aims to unlock the potential of modern immunotherapy in some of the most resistant "cold" tumors. This second narrative is currently being written in the clinic, representing a scientifically sound attempt to salvage a valuable asset by applying it to a new therapeutic paradigm.

9.2 Key Learnings from the Evofosfamide Development Program

The development history of Evofosfamide offers several critical lessons for the field of oncology drug development:

  1. The Primacy of Pharmaceutical Science: The MAESTRO trial failure is a stark reminder that even the most elegant biological mechanism can be undone by pharmaceutical and pharmacokinetic issues. It underscores the absolute necessity of conducting thorough bridging studies to ensure bioequivalence whenever a drug's formulation is altered, particularly between late-stage trials.
  2. The Challenge of Translating HAP Efficacy: The disconnect between the uniformly positive preclinical data and the mixed clinical results highlights the complexity of tumor hypoxia in humans. The heterogeneity of oxygen levels, metabolic activity, and reductase enzyme expression within and between patient tumors presents a significant challenge that bulk hypoxia measurements may fail to capture. Future HAP development will require more sophisticated, functional biomarkers to identify patients most likely to benefit.
  3. The Value of Re-evaluating "Failed" Assets: The revival of Evofosfamide demonstrates that a clinical trial failure does not always invalidate a drug's core mechanism. As scientific understanding evolves, particularly in areas like the TME and immuno-oncology, assets that failed under an old paradigm may find new and potent applications in a new one. A deep understanding of a drug's full biological effects, including those initially considered secondary, can unlock future value.

9.3 Concluding Remarks on Therapeutic Potential

Despite its challenging history, Evofosfamide possesses a unique and highly relevant mechanism of action. The problem it was designed to address—tumor hypoxia—has only grown in importance, now recognized as a central mediator of resistance to immunotherapy, the current pillar of cancer treatment. Its potential to functionally remodel the TME and sensitize immune-excluded tumors to checkpoint blockade is compelling and supported by emerging clinical evidence. The ongoing Phase 1/2 trial of Evofosfamide in combination with dual checkpoint blockade (NCT06782555) represents the definitive test of this new therapeutic hypothesis. Its outcome, anticipated around 2028, will ultimately determine whether the second narrative of Evofosfamide can conclude with the clinical success that has long been its promise.

Works cited

  1. Evofosfamide - Wikipedia, accessed September 13, 2025, https://en.wikipedia.org/wiki/Evofosfamide
  2. Evofosfamide: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed September 13, 2025, https://go.drugbank.com/drugs/DB06091
  3. Influence of the Hypoxia-Activated Prodrug Evofosfamide (TH-302) on Glycolytic Metabolism of Canine Glioma: A Potential Improvement in Cancer Metabolism. - eScholarship, accessed September 13, 2025, https://escholarship.org/uc/item/6xk056tp
  4. Influence of the Hypoxia-Activated Prodrug Evofosfamide (TH-302) on Glycolytic Metabolism of Canine Glioma - PubMed Central, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10705110/
  5. Evofosfamide Enhances Sensitivity of Breast Cancer Cells to Apoptosis and Natural-Killer-Cell-Mediated Cytotoxicity Under Hypoxic Conditions - MDPI, accessed September 13, 2025, https://www.mdpi.com/2072-6694/17/12/1988
  6. Hypoxia-Activated Prodrug Evofosfamide Treatment in Pancreatic Ductal Adenocarcinoma Xenografts Alters the Tumor Redox Status to Potentiate Radiotherapy - PMC, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8568781/
  7. Hypoxia-Activated Prodrug Evofosfamide Treatment in Pancreatic Ductal Adenocarcinoma Xenografts Alters the Tumor Redox Status to Potentiate Radiotherapy - PubMed, accessed September 13, 2025, https://pubmed.ncbi.nlm.nih.gov/32787454/
  8. Definition of evofosfamide - NCI Drug Dictionary - NCI, accessed September 13, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/evofosfamide
  9. Evofosfamide (TH-302) | Others | 99.82%(HPLC) | In Stock - Selleck Chemicals, accessed September 13, 2025, https://www.selleckchem.com/products/th-302.html
  10. Merck KGaA and Threshold Announce Global Agreement to Co-Develop and Commercialize Phase III Hypoxia-Targeted Drug TH-302 | Fierce Biotech, accessed September 13, 2025, https://www.fiercebiotech.com/biotech/merck-kgaa-and-threshold-announce-global-agreement-to-co-develop-and-commercialize-phase
  11. evofosfamide | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed September 13, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?tab=clinical&ligandId=8695
  12. Evofosfamide – Knowledge and References - Taylor & Francis, accessed September 13, 2025, https://taylorandfrancis.com/knowledge/Medicine_and_healthcare/Pharmaceutical_medicine/Evofosfamide/
  13. MAESTRO Trial Shows Evofosfamide Provides Little Benefit in STS - Targeted Oncology, accessed September 13, 2025, https://www.targetedonc.com/view/maestro-trial-shows-evofosfamide-provides-little-benefit-in-sts
  14. Merck abandons evofosfamide as two trials fail - PMLiVE, accessed September 13, 2025, https://pmlive.com/pharma_news/merck_abandons_evofosfamide_as_two_trials_fail_883775/
  15. Acquisition Strengthens “Cold” Tumor Targeting Pipeline - ImmunoGenesis, accessed September 13, 2025, https://www.immunogenesis.com/acquisition-strengthens-cold-tumor-targeting-pipeline/
  16. Evofosfamide Trial Targets Hypoxia in Resistant Solid Tumors, accessed September 13, 2025, https://www.targetedonc.com/view/evofosfamide-trial-targets-hypoxia-in-resistant-solid-tumors
  17. Evofosfamide | C9H16Br2N5O4P | CID 11984561 - PubChem, accessed September 13, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/11984561
  18. Evofosfamide - Drug Targets, Indications, Patents - Patsnap Synapse, accessed September 13, 2025, https://synapse.patsnap.com/drug/d6eac6d292e54529bd973811a2c75796
  19. Evofosfamide | TH-302 | CAS#918633-87-1 | MedKoo Biosciences, accessed September 13, 2025, https://www.medkoo.com/products/4926
  20. Evofosfamide (7507) by Tocris, Part of Bio-Techne, accessed September 13, 2025, https://www.bio-techne.com/p/small-molecules-peptides/evofosfamide_7507
  21. CAS 918633-87-1 Evofosfamide - BOC Sciences, accessed September 13, 2025, https://www.bocsci.com/evofosfamide-cas-918633-87-1-item-460516.html
  22. Compound: EVOFOSFAMIDE (CHEMBL260046) - ChEMBL - EMBL-EBI, accessed September 13, 2025, https://www.ebi.ac.uk/chembl/explore/compound/CHEMBL260046
  23. TH-302 (Evofosfamide, CAS Number: 918633-87-1) | Cayman Chemical, accessed September 13, 2025, https://www.caymanchem.com/product/22195/th-302
  24. evofosfamide | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed September 13, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=8695
  25. Cellular pharmacology of evofosfamide (TH-302): A critical re-evaluation of its bystander effects | Request PDF - ResearchGate, accessed September 13, 2025, https://www.researchgate.net/publication/327109762_Cellular_pharmacology_of_evofosfamide_TH-302_A_critical_re-evaluation_of_its_bystander_effects
  26. An Intratumor Pharmacokinetic/Pharmacodynamic Model for the Hypoxia-Activated Prodrug Evofosfamide (TH-302): Monotherapy Activity is Not Dependent on a Bystander Effect - PubMed Central, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6314220/
  27. An Intratumor Pharmacokinetic/Pharmacodynamic Model for the Hypoxia-Activated Prodrug Evofosfamide (TH-302): Monotherapy Activity is Not Dependent on a Bystander Effect - ResearchGate, accessed September 13, 2025, https://www.researchgate.net/publication/330075166_An_Intratumor_PharmacokineticPharmacodynamic_Model_for_the_Hypoxia-Activated_Prodrug_Evofosfamide_TH-302_Monotherapy_Activity_is_Not_Dependent_on_a_Bystander_Effect
  28. The Hypoxia-Activated Prodrug TH-302: Exploiting Hypoxia in Cancer Therapy - PMC, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8091515/
  29. Hypoxia-Targeting Drug Evofosfamide (TH-302) Enhances Sunitinib Activity in Neuroblastoma Xenograft Models - PMC, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6041570/
  30. Influence of the Hypoxia-Activated Prodrug Evofosfamide (TH-302) on Glycolytic Metabolism of Canine Glioma: A Potential Improvement in Cancer Metabolism - MDPI, accessed September 13, 2025, https://www.mdpi.com/2072-6694/15/23/5537
  31. Evofosfamide (TH-302) | Hypoxia-Activated Prodrug - MedchemExpress.com, accessed September 13, 2025, https://www.medchemexpress.com/TH-302.html
  32. Evofosfamide (TH-302) | CAS 918633-87-1 | InvivoChem, accessed September 13, 2025, https://invivochem.net/th-302.html
  33. Mechanism of evofosfamide action. (A) Structure of evofosfamide (also known as TH302). (B) After crossing the cell membrane, the nitro group of evofosfamide is reduced by 1e-reductases to generate the radical form of the drug. (C) Under normoxic conditions, this reaction is rapidly reversed to regenerate the original drug. (D) Under hypoxic conditions, this reaction is not reversed and the unstable radical fragments to generate the alkylating agent Br-IPM. (E) Br-IPM crosslinks DNA, targeting the cell for apoptosis or cell cycle arrest. Br-IPM, bromo-isophosphoramide. - ResearchGate, accessed September 13, 2025, https://www.researchgate.net/figure/Mechanism-of-evofosfamide-action-A-Structure-of-evofosfamide-also-known-as-TH302_fig1_343635675
  34. HNSCC (Head and Neck Squamous Cell Cancer Unknown Status Phase 1 Trials for Evofosfamide (DB06091) - DrugBank, accessed September 13, 2025, https://go.drugbank.com/indications/DBCOND0103106/clinical_trials/DB06091?phase=1&status=unknown_status
  35. Evofosfamide Terminated Phase 1 Trials for Pancreatic Cancer Treatment - DrugBank, accessed September 13, 2025, https://go.drugbank.com/drugs/DB06091/clinical_trials?conditions=DBCOND0028482&phase=1&purpose=treatment&status=terminated
  36. Evofosfamide (TH-302) in combination with gemcitabine in ..., accessed September 13, 2025, https://ascopubs.org/doi/10.1200/jco.2016.34.4_suppl.193
  37. MAESTRO: A randomized, double-blind phase III study of evofosfamide (Evo) in combination with gemcitabine (Gem) in previously untreated patients (pts) with metastatic or locally advanced unresectable pancreatic ductal adenocarcinoma (PDAC). | Journal of Clinical Oncology - ASCO Publications, accessed September 13, 2025, https://ascopubs.org/doi/10.1200/JCO.2016.34.15_suppl.4007
  38. Adding Evofosfamide to Doxorubicin in Advanced ... - The ASCO Post, accessed September 13, 2025, https://ascopost.com/News/57811
  39. Doxorubicin plus evofosfamide versus doxorubicin alone in locally ..., accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7771354/
  40. Phase I study of evofosfamide, an investigational hypoxia-activated ..., accessed September 13, 2025, https://pubmed.ncbi.nlm.nih.gov/27169385/
  41. A Phase 2 biomarker-enriched study of evofosfamide (TH-302) in patients with advanced melanoma. - ASCO, accessed September 13, 2025, https://www.asco.org/abstracts-presentations/ABSTRACT152326
  42. Immunotherapy Study of Evofosfamide in Combination With Ipilimumab | ClinicalTrials.gov, accessed September 13, 2025, https://clinicaltrials.gov/study/NCT03098160
  43. A Phase I Dose Escalation Study to Evaluate the Safety and Tolerability of Evofosfamide in Combination with Ipilimumab in Advanced Solid Malignancies, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8172466/
  44. Metastatic Prostate Cancer Recruiting Phase 1 / 2 Trials ... - DrugBank, accessed September 13, 2025, https://go.drugbank.com/indications/DBCOND0033188/clinical_trials/DB06091?phase=1%2C2&status=recruiting
  45. Unexpected pharmacokinetics of evofosfamide observed in phase III MAESTRO study., accessed September 13, 2025, https://www.asco.org/abstracts-presentations/ABSTRACT221667
  46. A Phase I Study of Evofosfamide, an Investigational Hypoxia-Activated Prodrug, in Patients with Advanced Leukemia, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4946992/
  47. Threshold Pharmaceuticals Reports Third Quarter Financial Results - SEC.gov, accessed September 13, 2025, https://www.sec.gov/Archives/edgar/data/1183765/000114420416131841/v452304_ex99-1.htm
  48. Evofosfamide fails in two late-stage cancer trials - European Pharmaceutical Review, accessed September 13, 2025, https://www.europeanpharmaceuticalreview.com/news/37091/evofosfamide-fails-in-two-late-stage-cancer-trials/
  49. Evofosfamide (TH-302) in Combination with Gemcitabine in Previously Untreated Patients with Metastatic or Locally Advanced Unresectable Pancreatic Ductal Adenocarcinoma: Primary Analysis of the Randomized, Double-Blind Phase III MAESTRO Study - ASCO, accessed September 13, 2025, https://www.asco.org/abstracts-presentations/ABSTRACT171197
  50. Comparison of hypoxia-activated prodrug evofosfamide (TH-302) and ifosfamide in preclinical non-small cell lung cancer models | Request PDF - ResearchGate, accessed September 13, 2025, https://www.researchgate.net/publication/292154161_Comparison_of_hypoxia-activated_prodrug_evofosfamide_TH-302_and_ifosfamide_in_preclinical_non-small_cell_lung_cancer_models
  51. Comparison of hypoxia-activated prodrug evofosfamide (TH-302) and ifosfamide in preclinical non-small cell lung cancer models - PMC, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5036787/
  52. Unexpected pharmacokinetics of evofosfamide observed in phase III ..., accessed September 13, 2025, https://ascopubs.org/doi/10.1200/JCO.2018.36.15_suppl.2568
  53. NCT06782555 | A Study of Evofosfamide in Combination with Zalifrelimab and Balstilimab, accessed September 13, 2025, https://www.clinicaltrials.gov/study/NCT06782555?term=AREA%5BBasicSearch%5D(th%20302)&rank=1
  54. 853 Evofosfomide potentiates the efficacy of a novel anti-PD-L1/PD-L2 monoclonal antibody against immune excluded tumors, accessed September 13, 2025, https://jitc.bmj.com/content/11/Suppl_1/A952

Published at: September 13, 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.