MedPath

Glufosfamide Advanced Drug Monograph

Published:Oct 17, 2025

Generic Name

Glufosfamide

Drug Type

Small Molecule

Chemical Formula

C10H21Cl2N2O7P

CAS Number

132682-98-5

Glufosfamide (DB06177): A Comprehensive Report on a Targeted Alkylating Agent

Executive Summary

[Glufosfamide (DrugBank ID: DB06177) is an investigational, third-generation oxazaphosphorine derivative designed as a targeted chemotherapeutic agent. Structurally, it is a conjugate of the active cytotoxic metabolite of ifosfamide—isophosphoramide mustard (IPM)—and a $\beta$-D-glucose molecule. This design is predicated on the hypothesis of metabolic targeting, aiming to exploit the increased glucose uptake characteristic of many malignant tumors to achieve preferential drug delivery and enhanced efficacy. The mechanism of action involves cellular uptake via glucose transporters, followed by intracellular cleavage to release the active IPM, which then functions as a DNA alkylating agent, inducing cell cycle arrest and apoptosis. A key theoretical advantage of this design is the circumvention of hepatic cytochrome P450 activation required by its parent compound, ifosfamide, thereby avoiding the production of toxic metabolites such as acrolein.]

[Despite its elegant scientific rationale, the clinical development of Glufosfamide has been protracted and challenging. Clinical trials, primarily focused on metastatic pancreatic cancer, have demonstrated modest efficacy signals but have been consistently hampered by a narrow therapeutic window. The drug's safety profile is dominated by significant, dose-limiting renal and hematologic toxicities. The very mechanism designed for tumor targeting—uptake by glucose transporters—is also responsible for high drug concentration in the renal tubules, leading to on-target, off-tumor nephrotoxicity. A pivotal Phase 3 trial in second-line pancreatic cancer conducted by Threshold Pharmaceuticals failed to meet its primary endpoint of improving overall survival in 2007. However, the drug's development has been revived by Eleison Pharmaceuticals, which has initiated a new Phase 3 trial (NCT01954992) with a modified design. The future of Glufosfamide is contingent on this ongoing trial and the potential identification of a specific patient subpopulation, defined by predictive biomarkers, in which its therapeutic benefits can decisively outweigh its inherent risks.]

1.0 Introduction to Glufosfamide: A Third-Generation Oxazaphosphorine Derivative

Glufosfamide is an investigational small molecule drug belonging to the oxazaphosphorine class of chemotherapeutics, representing a third-generation evolution of these alkylating agents.[1] It has been evaluated for the treatment of various solid tumors, including pancreatic, ovarian, lung, and soft tissue cancers.[1]

The therapeutic rationale behind Glufosfamide is rooted in a strategy of metabolic targeting. The molecule is a glycosidic conjugate, created by covalently bonding isophosphoramide mustard (IPM)—the active cytotoxic moiety of the widely used drug ifosfamide—to a $\beta$-D-glucose molecule.[5] This molecular design is intended to exploit a well-known metabolic hallmark of cancer known as the Warburg effect, wherein many tumor cells exhibit a significantly higher rate of glucose uptake and glycolysis compared to normal tissues.[9] The central hypothesis is that by "disguising" the cytotoxic agent as a glucose molecule, Glufosfamide can hijack the overexpressed glucose transport systems on cancer cells to achieve preferential accumulation within the tumor.[9] This targeted delivery is intended to increase the intratumoral concentration of the active drug, thereby enhancing its anti-cancer activity while potentially reducing the systemic toxicities associated with its parent compound, ifosfamide.[7]

2.0 Molecular Profile and Physicochemical Characteristics

[The precise identification and characterization of Glufosfamide are essential for research, development, and regulatory purposes. Its properties are well-documented across multiple chemical and pharmacological databases.]

Chemical Identity

[Glufosfamide is a distinct chemical entity with the following identifiers:]

  • DrugBank ID: DB06177 [13]
  • Type: Small Molecule [1]
  • CAS Number: 132682-98-5 [5]
  • Synonyms: The compound is known by several names, including D-19575, D 19575, Glucosylifosfamide mustard, $\beta$-D-Glc-IPM, glucophosphamide, and $\beta$-D-glucosylisophosphoramide mustard.[1]
  • IUPAC Name: N,N'-Bis(2-chloroethyl)phosphorodiamidic acid beta-D-glucopyranosyl ester.[6] A more systematic IUPAC name is (2S,3R,4S,5S,6R)-3,4,5-Trihydroxy-6-(hydroxymethyl)oxan-2-yl N,N′-bis(2-chloroethyl)phosphorodiamidate.[9]
  • Chemical Formula: $C_{10}H_{21}Cl_{2}N_{2}O_{7}P$.[5] One source reports a formula of $C_{7}H_{17}N_{2}O_{4}P$, which appears to be an error given the consistency of the $C_{10}$ formula across other chemical databases.[19]
  • Molecular Weight: 383.16 g/mol.[5] The value of 224.19 g/mol reported in one source is inconsistent with the correct chemical formula and is considered an outlier.[19]

Structural and Stereochemical Details

[The three-dimensional structure and stereochemistry are critical to the biological activity of Glufosfamide.]

  • SMILES Code: O=P(NCCCl)(NCCCl)O[C@H]1[C@@H]([C@H]([C@@H]([C@@H](CO)O1)O)O)O [5]
  • InChI Key: PSVUJBVBCOISSP-SPFKKGSWSA-N [6]
  • Stereochemistry: The biologically active compound is specifically the $\beta$-anomer, with the IPM moiety linked to D-glucose via a $\beta$-glycosidic bond.[19] An L-glucose isomer, designated D-23380, has also been synthesized and is available for research purposes, allowing for comparative studies of stereospecific uptake and activity.[19]

Physicochemical Properties

  • Appearance: White crystalline powder.[19]
  • Solubility: Glufosfamide is soluble in dimethyl sulfoxide (DMSO) and demonstrates high solubility in water, with one source reporting up to 200 mg/mL.[6]
  • Storage and Stability: For long-term storage, a temperature of -20°C is recommended.[5] Stock solutions prepared in DMSO should be stored in aliquots to avoid repeated freeze-thaw cycles and are typically stable for up to one month when stored at -20°C.[5]

Table 1: Key Identifiers and Physicochemical Properties of Glufosfamide

ParameterValueSource(s)
Identifiers
DrugBank IDDB0617713
CAS Number132682-98-55
IUPAC NameN,N'-Bis(2-chloroethyl)phosphorodiamidic acid beta-D-glucopyranosyl ester6
Chemical Properties
Chemical Formula$C_{10}H_{21}Cl_{2}N_{2}O_{7}P$5
Molecular Weight383.16 g/mol5
InChI KeyPSVUJBVBCOISSP-SPFKKGSWSA-N6
Physical Properties
AppearanceWhite crystalline powder19
SolubilitySoluble in water (200 mg/mL) and DMSO6
Storage-20°C (long-term)5

3.0 Mechanism of Action: A Strategy of Metabolic Targeting

[The mechanism of action of Glufosfamide is a direct consequence of its innovative dual-component molecular design, which combines a potent cytotoxic agent with a biological targeting moiety.]

Dual-Component Design

  • The Alkylating Moiety: The pharmacologically active component of Glufosfamide is isophosphoramide mustard (IPM).[5] IPM is the same active metabolite that is responsible for the therapeutic effects of the conventional chemotherapeutic drug ifosfamide. As a bifunctional alkylating agent, IPM exerts its cytotoxic effects by forming covalent bonds with nucleophilic sites on DNA bases. This leads to the formation of intra-strand and inter-strand DNA cross-links, which physically obstruct the DNA double helix. This damage disrupts critical cellular processes, including DNA replication and transcription, ultimately triggering cell cycle arrest and inducing apoptosis (programmed cell death).[1]
  • The Glucose Targeting Moiety: The $\beta$-D-glucose component is the key strategic element that differentiates Glufosfamide from its parent compound. It is designed to function as a "Trojan horse," enabling the drug to hijack the glucose transport machinery of cells for entry.[9]

Cellular Uptake and Activation

  • Transport: The cellular uptake of Glufosfamide is an active process mediated by glucose transporters, particularly sodium-dependent glucose co-transporters (SGLTs).[7] Research has specifically implicated SAAT1, a low-affinity sodium/glucose cotransporter, in this process, although other transporters may also contribute.[22] This transport mechanism is central to the drug's therapeutic rationale, as many aggressive cancers, including pancreatic cancer, are known to overexpress glucose transporters to satisfy their high metabolic demands for energy and anabolic precursors.[9]
  • Intracellular Release: Once Glufosfamide has been transported into the cell, the glycosidic linkage between the glucose molecule and the IPM is cleaved. This activation step is believed to be catalyzed by intracellular glucosidases, which are ubiquitous enzymes. This cleavage releases the active IPM directly within the target cell, where it can then exert its DNA-damaging effects.[7]

Key Advantages Over Ifosfamide

[The design of Glufosfamide offers two primary theoretical advantages over conventional ifosfamide therapy:]

  • Bypassing Hepatic Activation: Ifosfamide is a prodrug that is inert until it undergoes metabolic activation in the liver, a process mediated by the cytochrome P450 (CYP450) enzyme system. Glufosfamide, by contrast, is a pre-activated derivative, as it already contains the active IPM moiety.[7][ This circumvents the need for hepatic metabolism, potentially leading to a more consistent pharmacokinetic profile that is less susceptible to variations in liver enzyme activity.]
  • Reduced Systemic Toxicity: The hepatic metabolism of ifosfamide generates several toxic byproducts. The most notable are acrolein, which is responsible for hemorrhagic cystitis (a severe form of bladder inflammation), and chloroacetaldehyde, which is associated with neurotoxicity.[7][ Because Glufosfamide's activation pathway does not involve the same metabolic steps, it does not produce these toxic metabolites, providing a strong mechanistic basis for a potentially improved safety profile.]

This elegant mechanism of targeting, however, presents a fundamental challenge. The very biological process exploited for tumor selectivity is also the direct cause of the drug's principal toxicity. The kidneys, particularly the cells of the proximal tubules, are responsible for reabsorbing glucose from the glomerular filtrate to prevent its loss in urine. This physiological function is mediated by a high density of the same sodium-glucose cotransporters that Glufosfamide targets. Consequently, the drug becomes highly concentrated in these renal cells. Following uptake, intracellular cleavage releases the potent alkylating agent IPM, causing direct cellular damage. This leads to the clinically observed nephrotoxicity, including renal tubular acidosis and, in severe cases, renal failure.[22][ This creates an on-target, off-tumor toxicity that is mechanistically inseparable from its intended anti-cancer action, resulting in an inherently narrow therapeutic window that has proven to be a major obstacle throughout its clinical development.]

4.0 Preclinical and Clinical Pharmacokinetic Profile

[The pharmacokinetic properties of Glufosfamide—its absorption, distribution, metabolism, and excretion (ADME)—have been characterized in both preclinical models and human clinical trials, providing a clear picture of its behavior in the body.]

Administration and Distribution

Glufosfamide is administered intravenously, with clinical trial protocols utilizing infusion durations ranging from one to six hours.[23] Preclinical studies in animal models demonstrated favorable distribution characteristics, including high tissue distribution and low binding to plasma proteins.[22] This is consistent with findings from human studies where analysis of plasma samples using Phosphorus-31 Nuclear Magnetic Resonance ($^{31}$P NMR) revealed that unchanged Glufosfamide was the only drug-related phosphorus-containing compound detected systemically. This indicates that the drug circulates intact as the prodrug prior to its uptake into tissues or its elimination by the kidneys.[30]

Metabolism

A central feature of Glufosfamide's design is that it does not require hepatic activation by the CYP450 system, unlike its parent compound ifosfamide.[7] Its metabolism is believed to occur intracellularly following transport into cells. Clinical pharmacokinetic data suggest that this metabolic process is capacity-limited. At higher doses (e.g., 8 g/m²), a slight increase in the elimination half-life ($t_{1/2}$) and a corresponding decrease in plasma clearance ($CL_p$) were observed compared to lower dose levels (4.5 to 6 g/m²). This non-linear pharmacokinetic behavior implies that the enzymes responsible for its intracellular cleavage, likely glucosidases, can become saturated at high drug concentrations.[30]

Excretion

The primary route of elimination for Glufosfamide and its metabolites is renal excretion. Preclinical models showed rapid clearance by the kidneys.[22][ Detailed human excretion studies using $^{31}$P NMR have provided a quantitative understanding of this process:]

  • Between 30% and 60% of the total administered dose is recovered in the urine within a 24-hour period. The proportion of the dose excreted increases as the administered dose increases.[30]
  • The vast majority of the drug excreted in the urine is in the form of unchanged Glufosfamide, which accounts for 83% to 89% of all phosphorus-containing compounds detected.[30]
  • Metabolites, including the active IPM and its degradation products phosphorylethanolamine (PEA) and glycerophosphorylethanolamine (GPE), are only minor components in the urine, with IPM and PEA each accounting for just 2-3% of the total excreted compounds.[30]
  • The excretion process is rapid, with approximately 84-86% of the total 24-hour urinary excretion occurring within the first 8 hours following the start of the infusion.[30]

Pharmacokinetic Interactions

In clinical trials where Glufosfamide was administered in combination with gemcitabine, pharmacokinetic analyses were conducted to assess potential drug-drug interactions. These studies concluded that there was no significant pharmacokinetic interaction between the two agents, meaning that neither drug substantially altered the clearance or exposure of the other.[23][ This finding is clinically important, as it simplifies the dosing of the combination regimen and reduces the risk of unexpected toxicity arising from altered drug metabolism or elimination.]

The pharmacokinetic profile provides strong corroborating evidence for the drug's mechanism of action and its associated toxicity. The fact that the majority of the drug is rapidly cleared by the kidneys and excreted in the urine as the unchanged parent compound demonstrates that a large amount of the intact prodrug is presented to the renal tubules.[30][ This high renal exposure, coupled with the efficient uptake of the glucose-conjugated drug by SGLTs in the tubular cells, offers a clear pharmacokinetic basis for the observed nephrotoxicity. The kidneys are not merely a passive route of clearance; they actively concentrate the drug, leading to localized, high-level exposure of the renal tissue to the cytotoxic IPM upon intracellular cleavage.]

5.0 Clinical Development Program: A History of Trials and Tribulations

[The clinical investigation of Glufosfamide has spanned more than two decades, involving multiple corporate sponsors and exploring several solid tumor indications. The primary focus has consistently been on pancreatic cancer, an area of high unmet medical need. The drug's journey has been characterized by early signs of promise, a major setback in a pivotal trial, and a recent strategic revival of its development program.]

Table 2: Summary of Major Clinical Trials for Glufosfamide

Trial IdentifierPhaseIndication(s)Sponsor(s)StatusKey Design / Outcome
NCT000992943Metastatic Pancreatic Cancer (2nd Line)Threshold PharmaceuticalsCompletedRandomized trial of Glufosfamide + BSC vs. BSC alone. Failed to meet primary endpoint of overall survival (OS).32
NCT019549923Metastatic Pancreatic Cancer (2nd Line)Eleison PharmaceuticalsRecruitingRandomized trial of Glufosfamide vs. 5-FU in gemcitabine-pretreated patients. Primary endpoint is OS.1
NCT001027521/2Advanced Solid Tumors / Pancreatic Cancer (1st Line)Threshold PharmaceuticalsCompletedDose-escalation study of Glufosfamide + Gemcitabine. Established MTD and showed promising activity in pancreatic cancer.14
NCT004414672Advanced Soft Tissue SarcomaEleison PharmaceuticalsCompletedOpen-label study in pre-treated patients. Showed some clinical activity but was marked by severe renal toxicity.1
NCT004425982Ovarian CancerEleison PharmaceuticalsTerminatedRandomized study of two dosing schedules. Terminated due to lack of efficacy and enrollment difficulties related to poor renal function.1
NCT004355782Small Cell Lung CarcinomaThreshold PharmaceuticalsTerminatedOpen-label study in recurrent sensitive SCLC. Terminated early for reasons not fully detailed.1
NCT000050532Advanced Pancreatic CancerEORTCCompletedRandomized trial evaluating Glufosfamide with or without hydration.4

5.1 Pancreatic Cancer: The Primary Therapeutic Focus

5.1.1 Early Phase Evaluation and Combination Therapy (NCT00102752, NCT00005053)

Initial clinical studies sought to establish the safety, tolerability, and optimal dose of Glufosfamide, both as a single agent and in combination. A key Phase 1/2 dose-escalation study (NCT00102752) evaluated Glufosfamide combined with the standard-of-care agent gemcitabine in patients with advanced solid tumors, including a cohort with pancreatic cancer.[14] This trial successfully established that the full dose of Glufosfamide (4,500 mg/m²) could be safely administered with gemcitabine.[23]

The subsequent Phase 2 portion of this study focused on 29 chemotherapy-naïve patients with advanced pancreatic adenocarcinoma.[11][ The results demonstrated promising anti-tumor activity:]

  • Efficacy: The confirmed partial response (PR) rate was 18% (5 of 28 evaluable patients), with a median response duration of 8.4 months. An additional 39% of patients achieved stable disease (SD). The median progression-free survival (PFS) was 3.7 months, and the median overall survival (OS) was 6.0 months. Notably, the 1-year survival rate was 32%, which was considered encouraging in this disease setting.[12]
  • Toxicity: This efficacy came at the cost of significant toxicity. The combination therapy led to pronounced hematologic side effects, with Grade 3/4 neutropenia occurring in 79% of patients. Renal toxicity was also a major concern; creatinine clearance (CrCL) fell below the safety threshold of 60 mL/min in 37% of patients, and four patients developed renal failure.[12]

5.1.2 The First Pivotal Trial (NCT00099294): A Statistically Insignificant Outcome

Based on the early signs of activity, Threshold Pharmaceuticals advanced Glufosfamide into a pivotal Phase 3 trial (NCT00099294). This multinational, randomized study enrolled 303 patients with metastatic pancreatic cancer who had progressed after prior gemcitabine-based therapy. Patients were randomized to receive either Glufosfamide plus Best Supportive Care (BSC) or BSC alone.[4]

  • Primary Endpoint Failure: In February 2007, it was announced that the trial did not meet its primary endpoint of a statistically significant improvement in overall survival.[6]
  • Efficacy Data: While there was a trend favoring the investigational arm, it did not reach statistical significance. The median OS for patients treated with Glufosfamide was 105 days, compared to 84 days for those receiving BSC alone, representing an 18% improvement (Hazard Ratio 0.85; 95% Confidence Interval [CI] 0.66–1.08; p=0.19).[32]
  • Conclusion: The investigators concluded that Glufosfamide demonstrated low activity in this highly refractory patient population, and this trial failure represented a major setback for the drug's development program.[32]

5.1.3 The Relaunch: Eleison's Pivotal Program (NCT01954992)

Despite the previous Phase 3 failure, the development of Glufosfamide was later revived by Eleison Pharmaceuticals, which initiated a new pivotal Phase 3 trial (NCT01954992).[1]

  • Study Design: This ongoing trial is a large, randomized, open-label study designed to enroll approximately 480 patients with metastatic pancreatic adenocarcinoma who have previously been treated with gemcitabine. It compares Glufosfamide to an active comparator, fluorouracil (5-FU), which is a recognized treatment option in this setting. The primary endpoint remains overall survival.[29]
  • Current Status: The trial is actively recruiting patients at sites in the United States and is planned for expansion to Europe and Asia. The estimated primary completion date is in 2026, with final study completion anticipated in 2027.[1]

The decision to launch a second Phase 3 trial reflects a strategic re-evaluation of the drug. A key driver for this revival may have been a post-hoc subgroup analysis from the failed NCT00099294 trial. This analysis revealed a surprisingly large survival benefit in a very small subgroup of diabetic patients who were taking glucose-lowering agents, with a median OS of 13.7 months for those on Glufosfamide versus 2.4 months for those on BSC.[39] While not statistically robust due to the small numbers, this finding provided a tantalizing link to the drug's glucose-based mechanism. The design of the new trial (NCT01954992) explicitly excludes patients who are insulin-taking diabetics, a critical modification aimed at refining the patient population.[35] This, combined with a recent collaboration between Eleison and BullFrog AI to use artificial intelligence to analyze clinical data and identify predictive biomarkers, indicates a strategic shift from a broad approach to a more targeted, biomarker-driven one.[1][ This new strategy attempts to salvage the drug by precisely identifying the patient niche where its unique mechanism offers a clear advantage that outweighs its risks.]

5.2 Exploratory Studies in Other Solid Tumors

5.2.1 Soft Tissue Sarcoma (NCT00441467)

A Phase 2 study evaluated Glufosfamide in 22 patients with advanced, pre-treated soft tissue sarcoma.[1] The trial demonstrated a clinical benefit rate (defined as stable disease or better) of 36%. However, the study was marred by a high rate of severe renal toxicity. Five patients (23%) experienced renal failure, including one grade 5 (fatal) event. The investigators noted that the renal toxicity was higher than observed in other Glufosfamide studies and concluded that alternative dosing regimens would be necessary to improve the drug's therapeutic index in this population.[26]

5.2.2 Discontinued Programs: Ovarian and Small Cell Lung Cancer

  • Ovarian Cancer (NCT00442598): A Phase 2 trial in patients with platinum-resistant ovarian cancer was terminated prematurely.[1] The decision was based on a combination of a lack of observed efficacy and significant challenges with patient enrollment. A key issue was that many otherwise eligible patients, having received multiple prior lines of chemotherapy, had pre-existing renal impairment that made them ineligible for treatment with a potentially nephrotoxic agent.[37]
  • Small Cell Lung Cancer (SCLC) (NCT00435578): A Phase 2 trial designed to evaluate Glufosfamide in patients with recurrent, sensitive SCLC was also terminated early, though the specific reasons are less clearly detailed.[1]

6.0 Comprehensive Safety and Tolerability Assessment

[The safety profile of Glufosfamide has been extensively characterized across numerous clinical trials. A consistent pattern of dose-limiting toxicities (DLTs) has emerged, defining the drug's narrow therapeutic window and presenting the primary challenge to its clinical utility.]

Dose-Limiting Toxicities (DLTs)

[The clinical development of Glufosfamide has been consistently constrained by two major categories of dose-limiting toxicity:]

  • Renal Toxicity: This is the most significant and characteristic toxicity of Glufosfamide. It manifests across a spectrum of severity, including reversible renal tubular acidosis, elevations in serum creatinine, dose-dependent decreases in creatinine clearance (CrCL), and, in the most severe cases, acute renal failure.[22] This nephrotoxicity was identified as a DLT in early Phase 1 studies [22], was a pronounced issue in the Phase 2 combination trial with gemcitabine (where renal failure occurred in 4 of 29 patients) [25], and was the most common severe adverse event in the soft tissue sarcoma trial, where it was implicated in one patient's death.[26]
  • Hematologic Toxicity: Myelosuppression is the second major DLT. This primarily involves Grade 3 and Grade 4 neutropenia (a low count of neutrophils, a type of white blood cell) and thrombocytopenia (a low platelet count).[22] The incidence of severe hematologic toxicity was particularly high in the combination study with gemcitabine, a known myelosuppressive agent.[12]

Common Adverse Events

[Beyond the dose-limiting toxicities, the most frequently reported adverse events are generally of lower grade and are typical for cytotoxic chemotherapy:]

  • Gastrointestinal Effects: Nausea and vomiting are very common.[26]
  • Constitutional Symptoms: Fatigue is also frequently reported by patients.[26]

[The consolidated safety data across different clinical settings underscores that renal and hematologic toxicities are intrinsic properties of Glufosfamide, observed in both monotherapy and combination regimens.]

Table 3: Consolidated Safety Profile of Glufosfamide Across Key Trials

Adverse Event (Grade 3/4)Incidence in Pancreatic Cancer Trial (NCT00102752, Glufosfamide + Gemcitabine)Incidence in Sarcoma Trial (NCT00441467, Glufosfamide Monotherapy)Source(s)
Renal Toxicity
Renal Failure14% (4/29 patients)23% (5/22 patients, grades 3-5)25
Decreased CrCL (<60 mL/min)37% (10/27 patients)N/A25
Hematologic Toxicity
Neutropenia79% (23/29 patients)9% (2/22 patients)25
Thrombocytopenia34% (10/29 patients)5% (1/22 patients)25

7.0 Corporate and Regulatory History

[The development of Glufosfamide has been passed between several pharmaceutical companies over more than two decades, a journey that reflects both the drug's perceived potential and the significant hurdles encountered in its clinical evaluation.]

Chronological Development Pathway

  • Origins: Glufosfamide was first conceived and synthesized through a research collaboration between Asta Medica (a subsidiary of Degussa) and the prestigious German Cancer Research Centre (DKFZ) in Heidelberg, Germany.[21]
  • Baxter Oncology: In October 2001, Baxter International acquired the oncology division of Asta Medica, which was subsequently renamed Baxter Oncology GmbH.[16] Under Baxter's stewardship, Glufosfamide was advanced into Phase 2 clinical trials. However, in its 2002 Annual Report, Baxter announced the termination of the drug's development.[21] While the specific rationale for this decision was not publicly detailed, it occurred during a financially challenging period for the company.[44]
  • Threshold Pharmaceuticals: In August 2003, Threshold Pharmaceuticals acquired the exclusive rights to develop and commercialize Glufosfamide through a licensing agreement with Baxter.[21] Threshold became the primary driver of the drug's clinical program for nearly a decade, sponsoring multiple Phase 2 studies and the pivotal Phase 3 trial in pancreatic cancer (NCT00099294). The failure of this trial to meet its primary endpoint in 2007 was a major setback, leading Threshold to discontinue development in several indications and shift its corporate focus to other pipeline assets.[33]
  • Eleison Pharmaceuticals: The program was later acquired by Eleison Pharmaceuticals, the current sponsor.[1] Eleison has revived the development of Glufosfamide, initiating the new, ongoing Phase 3 trial (NCT01954992) in second-line pancreatic cancer, signaling a renewed belief in the drug's potential within a more carefully selected patient population.[35]

Key Regulatory Milestones

[Despite its challenging clinical history, Glufosfamide has received several important regulatory designations from health authorities in the United States and Europe, acknowledging the high unmet need in pancreatic cancer.]

  • U.S. Food and Drug Administration (FDA):
  • In November 2004, the FDA granted Fast Track status to Glufosfamide for the treatment of patients with metastatic pancreatic cancer refractory to gemcitabine.[21][ This designation is intended to facilitate the development and expedite the review of drugs that treat serious conditions and fill an unmet medical need.]
  • On September 18, 2006, the FDA granted Orphan Drug Designation to Glufosfamide for the treatment of pancreatic cancer.[45][ This status provides incentives, including market exclusivity and tax credits, to encourage the development of drugs for rare diseases affecting fewer than 200,000 people in the U.S.]
  • European Medicines Agency (EMA):
  • On April 15, 2011, the European Commission, upon the recommendation of the EMA's Committee for Orphan Medicinal Products (COMP), granted Orphan Drug Designation to Glufosfamide for the treatment of pancreatic cancer.[41] The designation was based on the life-threatening nature of the disease, its prevalence of approximately 1.3 in 10,000 people in the EU, and sufficient evidence suggesting that Glufosfamide might offer a significant benefit over existing therapies.[47]

8.0 Synthesis, Critical Analysis, and Future Outlook

[The multi-decade development history of Glufosfamide presents a compelling narrative of scientific innovation confronting biological complexity. The drug was born from an elegant and powerful rationale: leveraging the unique metabolic properties of cancer cells for targeted drug delivery. This concept of metabolic targeting remains a highly attractive strategy in oncology. However, the clinical translation of this concept has been fraught with challenges, primarily stemming from a narrow therapeutic index defined by on-target, off-tumor toxicity.]

[The central paradox of Glufosfamide is that its primary strength is also its greatest weakness. The drug appears to work precisely as designed, utilizing glucose transporters for cellular entry. This mechanism, intended to concentrate the cytotoxic payload in glucose-avid tumors, also leads to its accumulation in healthy renal tubular cells, which are physiologically rich in the same transporters. The resulting nephrotoxicity is therefore not an unexpected side effect but an inherent consequence of the drug's mechanism of action. This has consistently limited the dose that can be safely administered, likely preventing the achievement of a sufficiently high therapeutic concentration in tumors to produce robust and consistent efficacy across a broad patient population.]

[The future of Glufosfamide now rests entirely on the outcome of the ongoing Phase 3 trial, NCT01954992. This trial is not merely a repeat of the failed 2007 study but represents a strategic evolution toward a precision medicine approach. By modifying the study population and partnering with an AI firm to identify predictive biomarkers, Eleison Pharmaceuticals is attempting to define a specific patient subgroup where the efficacy signal is strong enough to justify the significant toxicity risks. Success in this trial would validate this biomarker-driven strategy and could finally carve out a clinical niche for Glufosfamide in the treatment of pancreatic cancer. Failure would likely mark the end of the drug's long and arduous journey.]

[Ultimately, the story of Glufosfamide serves as an important case study in modern drug development. It highlights the immense gap that can exist between a compelling preclinical hypothesis and clinical reality. It demonstrates how corporate strategy and perseverance can give a drug multiple chances at success. Most importantly, it underscores the critical shift in oncology from a "one-size-fits-all" paradigm to a more nuanced approach, where the key to unlocking a drug's potential may lie not in treating all patients, but in identifying the right patients. The final chapter for Glufosfamide will determine whether it becomes a valuable therapeutic agent for a select few or a cautionary tale of a rational design ultimately foiled by inescapable biology.]

Works cited

  1. Glufosfamide - Drug Targets, Indications, Patents - Patsnap Synapse, accessed October 17, 2025, https://synapse.patsnap.com/drug/7cf48863af1940ab966dd35173eb64c1
  2. Glufosfamide as a New Oxazaphosphorine Anticancer Agent - PubMed, accessed October 17, 2025, https://pubmed.ncbi.nlm.nih.gov/21427562/
  3. BullFrog AI and Eleison Pharmaceuticals Enter Agreement to Collaborate to Optimize Pivotal Phase 3 Trial, accessed October 17, 2025, https://ir.bullfrogai.com/news-events/press-releases/detail/59/bullfrog-ai-and-eleison-pharmaceuticals-enter-agreement-to-collaborate-to-optimize-pivotal-phase-3-trial
  4. Glufosfamide - MedPath, accessed October 17, 2025, https://trial.medpath.com/drug/a732a8abe9c63790/glufosfamide
  5. GLUFOSFAMIDE (D 19575) | CAS NO.:132682-98-5 | GlpBio, accessed October 17, 2025, https://www.glpbio.com/glufosfamide-d-19575.html
  6. Glufosfamide | CAS#132682-98-5 | DNA alkylating agent - MedKoo Biosciences, accessed October 17, 2025, https://www.medkoo.com/products/5495
  7. A Novel Alkylating Agent, Glufosfamide, Enhances the Activity of Gemcitabine In vitro and In vivo - PMC, accessed October 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC1950432/
  8. A Novel Alkylating Agent, Glufosfamide, Enhances the Activity of ..., accessed October 17, 2025, https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1950432/
  9. Glufosfamide - Wikipedia, accessed October 17, 2025, https://en.wikipedia.org/wiki/Glufosfamide
  10. What is Glufosfamide used for? - Patsnap Synapse, accessed October 17, 2025, https://synapse.patsnap.com/article/what-is-glufosfamide-used-for
  11. Glufosfamide (GLU) plus gemcitabine (GEM) in pancreatic adenocarcinoma: Preliminary results of a phase 2 trial - ASCO Publications, accessed October 17, 2025, https://ascopubs.org/doi/10.1200/jco.2007.25.18_suppl.15005
  12. PRESS RELEASE: Threshold Pharmaceuticals Positive Glufosfamide Results From a Phase 2 Clinical Trial | Fierce Biotech, accessed October 17, 2025, https://www.fiercebiotech.com/biotech/press-release-threshold-pharmaceuticals-positive-glufosfamide-results-from-a-phase-2
  13. DrugMapper, accessed October 17, 2025, https://idaapm.helsinki.fi/CompoundInfo?compound_id=2126862
  14. Glufosfamide Completed Phase 1 / 2 Trials for Pancreatic Neoplasms / Neoplasms Treatment | DrugBank Online, accessed October 17, 2025, https://go.drugbank.com/drugs/DB06177/clinical_trials?conditions=DBCOND0027948%2CDBCOND0028305&phase=1%2C2&purpose=treatment&status=completed
  15. CNP0074800.1: Glufosfamide - COCONUT, accessed October 17, 2025, https://coconut.naturalproducts.net/compounds/CNP0074800.1
  16. Glufosfamide (Baxter Oncology) - PubMed, accessed October 17, 2025, https://pubmed.ncbi.nlm.nih.gov/12431031/
  17. Glufosfamide - Eleison Pharmaceuticals - AdisInsight - Springer, accessed October 17, 2025, https://adisinsight.springer.com/drugs/800003258
  18. 132682-98-5 | MFCD00914610 | Glufosfamide - Aaronchem, accessed October 17, 2025, https://www.aaronchem.com/132682-98-5
  19. Glufosfamide (CAS 132682-98-5) - Niomech, accessed October 17, 2025, https://www.niomech.com/glufosfamide_mobile
  20. en.wikipedia.org, accessed October 17, 2025, https://en.wikipedia.org/wiki/Glufosfamide#:~:text=Glufosfamide%20utilizes%20the%20normal%20cell,own%20transport%20into%20the%20cell.
  21. Glufosfamide: beta-D-Glc-IPM, D 19575 - PubMed, accessed October 17, 2025, https://pubmed.ncbi.nlm.nih.gov/15801867/
  22. Phase I Trial of 6-Hour Infusion of Glufosfamide, a New Alkylating Agent With Potentially Enhanced Selectivity for Tumors That Overexpress Transmembrane Glucose Transporters: A Study of the European Organization for Research and Treatment of Cancer Early Clinical Studies Group - ASCO Publications, accessed October 17, 2025, https://ascopubs.org/doi/10.1200/JCO.2000.18.20.3535
  23. Glufosfamide: can we improve the process of anticancer agent development?, accessed October 17, 2025, https://www.semanticscholar.org/paper/da60ce64d0fb47b24a8141821fa27f40efa64866
  24. Glufosfamide: can we improve the process of anticancer agent development? - PubMed, accessed October 17, 2025, https://pubmed.ncbi.nlm.nih.gov/22424104/
  25. A phase 2 trial of glufosfamide in combination with gemcitabine in chemotherapy-naive pancreatic adenocarcinoma - PubMed, accessed October 17, 2025, https://pubmed.ncbi.nlm.nih.gov/19687729/
  26. Preliminary efficacy and safety results of glufosfamide (GLU) in ..., accessed October 17, 2025, https://ascopubs.org/doi/10.1200/jco.2008.26.15_suppl.10591
  27. Study Details | NCT00005053 | Glufosfamide With or Without Hydration in Treating Patients With Advanced Pancreatic Cancer | ClinicalTrials.gov, accessed October 17, 2025, https://www.clinicaltrials.gov/study/NCT00005053
  28. Phase I clinical and pharmacokinetic study of the glucose-conjugated cytotoxic agent D-19575 (glufosfamide) in patients with solid tumors - PubMed, accessed October 17, 2025, https://pubmed.ncbi.nlm.nih.gov/19479254/
  29. Study Details | NCT01954992 | Glufosfamide Versus 5-FU in Second Line Metastatic Pancreatic Cancer | ClinicalTrials.gov, accessed October 17, 2025, https://www.clinicaltrials.gov/study/NCT01954992
  30. Glufosfamide metabolism in patients using phosphorus-31 NMR ..., accessed October 17, 2025, https://aacrjournals.org/cancerres/article/64/7_Supplement/260/512580/Glufosfamide-metabolism-in-patients-using
  31. Glufosfamide (GLU) plus gemcitabine (GEM) in advanced solid tumors: Phase 1 results, accessed October 17, 2025, https://ascopubs.org/doi/10.1200/jco.2006.24.18_suppl.14022
  32. A randomised Phase III trial of glufosfamide compared with best supportive care in metastatic pancreatic adenocarcinoma previously treated with gemcitabine - PubMed, accessed October 17, 2025, https://pubmed.ncbi.nlm.nih.gov/19188061/
  33. Press Release: Threshold Pharmaceuticals Announces that Trial of Glufosfamide Misses Endpoint | Fierce Biotech, accessed October 17, 2025, https://www.fiercebiotech.com/biotech/press-release-threshold-pharmaceuticals-announces-trial-of-glufosfamide-misses-endpoint
  34. Glufosfamide Completed Phase 3 Trials for Pancreatic Cancer Treatment | DrugBank Online, accessed October 17, 2025, https://go.drugbank.com/drugs/DB06177/clinical_trials?conditions=DBCOND0028482&phase=3&purpose=treatment&status=completed
  35. Clinical Trials - Eleison Pharmaceuticals, accessed October 17, 2025, https://eleison-pharma.com/clinical-trials/
  36. NCT00102752 | Study of Safety and Efficacy of Glufosfamide in Combination With Gemcitabine in Cancer | ClinicalTrials.gov, accessed October 17, 2025, https://clinicaltrials.gov/study/NCT00102752
  37. Threshold Updates Glufosfamide Clinical Development Program - Fierce Biotech, accessed October 17, 2025, https://www.fiercebiotech.com/biotech/threshold-updates-glufosfamide-clinical-development-program
  38. Safety and Efficacy Study of Glufosfamide in Ovarian Cancer | ClinicalTrials.gov, accessed October 17, 2025, https://clinicaltrials.gov/study/NCT00442598
  39. Threshold Pharmaceuticals, Inc. Glufosfamide Results From Phase 3 Clinical Trial Presented at European Cancer Conference (ECCO) - BioSpace, accessed October 17, 2025, https://www.biospace.com/threshold-pharmaceuticals-inc-glufosfamide-results-from-phase-3-clinical-trial-presented-at-european-cancer-conference-ecco
  40. Glufosfamide is dead, long live glufosfamide! - Citeline News & Insights, accessed October 17, 2025, https://insights.citeline.com/SC023032/Glufosfamide-is-dead-long-live-glufosfamide/
  41. Our Pipeline - Eleison Pharmaceuticals, accessed October 17, 2025, https://eleison-pharma.com/pipeline/
  42. BullFrog AI partners with Eleison to enhance Phase III trial, accessed October 17, 2025, https://www.clinicaltrialsarena.com/news/bullfrog-eleison-trial/
  43. A Phase 1 dose-escalation trial of glufosfamide in combination with gemcitabine in solid tumors including pancreatic adenocarcinoma - PubMed, accessed October 17, 2025, https://pubmed.ncbi.nlm.nih.gov/17661038/
  44. Baxter Annual Report 2002, accessed October 17, 2025, https://www.annualreports.com/HostedData/AnnualReportArchive/b/NYSE_BAX_2002.pdf
  45. Threshold Pharmaceuticals, Inc. Receives Orphan Drug Designation For Glufosfamide, accessed October 17, 2025, https://www.biospace.com/threshold-pharmaceuticals-inc-receives-orphan-drug-designation-for-glufosfamide
  46. Glufosfamide - Orphanet, accessed October 17, 2025, https://www.orpha.net/en/drug/substance/252227?name=&mode=pat®ion=&status=all
  47. EU/3/11/851 - orphan designation for treatment of pancreatic cancer, accessed October 17, 2025, https://www.ema.europa.eu/en/medicines/human/orphan-designations/eu-3-11-851

Published at: October 17, 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.