MedPath

Perifosine Advanced Drug Monograph

Published:Aug 25, 2025

Generic Name

Perifosine

Drug Type

Small Molecule

Chemical Formula

C25H52NO4P

CAS Number

157716-52-4

Perifosine (DB06641): A Comprehensive Monograph on a Novel Akt Inhibitor from Preclinical Promise to Phase III Discontinuation

Executive Summary

Perifosine (DB06641) represents a significant and cautionary chapter in the history of targeted cancer therapy. Developed as a novel, first-in-class, orally bioavailable alkylphospholipid, it was designed to inhibit the serine/threonine kinase Akt, a central node in the PI3K/Akt/mTOR signaling pathway, which is frequently dysregulated in human cancers and is a critical mediator of cell survival, proliferation, and therapeutic resistance. The initial scientific rationale for Perifosine was compelling, targeting a high-value oncogenic driver with a unique, allosteric mechanism of action that distinguished it from conventional ATP-competitive kinase inhibitors.

Preclinical investigations revealed potent antiproliferative and pro-apoptotic activity across a broad spectrum of cancer cell lines and in vivo xenograft models. This promising laboratory data was subsequently bolstered by early-phase clinical trials. Notably, a randomized Phase II study in metastatic colorectal cancer (mCRC) demonstrated a dramatic and statistically significant improvement in both progression-free and overall survival when Perifosine was added to capecitabine. Similarly, Phase I/II trials in relapsed/refractory multiple myeloma (MM) showed impressive response rates and survival outcomes for the combination of Perifosine with bortezomib and dexamethasone. These encouraging results led to the initiation of large, pivotal Phase III registration trials for both indications, accompanied by Fast Track and Orphan Drug designations from regulatory authorities.

However, the trajectory of Perifosine took a decisive downturn at this late stage. In 2012, the Phase III X-PECT trial in mCRC was halted after it became clear that the addition of Perifosine offered no survival benefit over capecitabine alone, completely failing to replicate the robust efficacy signal seen in Phase II. This was followed in 2013 by the discontinuation of the Phase III trial in MM, which also failed to meet its primary endpoint of improving progression-free survival. These definitive failures led to the complete cessation of Perifosine's clinical development.

This report provides an exhaustive analysis of Perifosine, from its fundamental chemical properties and multifaceted mechanism of action to a critical review of its clinical trial performance. The narrative of Perifosine serves as a crucial case study in drug development, illustrating the profound challenges of translating early-phase clinical success into late-stage confirmation. Its history underscores the complexities of targeting central signaling pathways, the imperative for validated predictive biomarkers in patient selection, and the potential pitfalls of promising but ultimately deceptive pharmacokinetic profiles and small, statistically fragile early-phase trials.

Drug Profile and Chemical Characteristics

A precise understanding of Perifosine's chemical identity and properties is fundamental to appreciating its unique pharmacological profile and the rationale behind its development.

Identification and Nomenclature

Perifosine is a synthetically derived small molecule compound that has been extensively cataloged under various identifiers across chemical and pharmacological databases. Establishing this nomenclature is the first step in a comprehensive review.

  • Generic Name: Perifosine [1]
  • DrugBank Accession Number: DB06641 [1]
  • CAS Number: 157716-52-4 [1]
  • Type/Modality: Small Molecule [1]
  • Synonyms and External IDs: Throughout its development and investigation, Perifosine has been referred to by several alternative names and codes. These include the developmental codes KRX-0401 and D 21266, as well as the National Cancer Institute (NCI) identifier NSC-639966.[1]

Physicochemical Properties

The specific molecular structure and physical characteristics of Perifosine define its behavior both in vitro and in vivo, directly influencing its mechanism of action, formulation, and pharmacokinetic profile. The key properties are summarized in Table 1.

The structure of Perifosine is amphipathic, featuring a long, nonpolar octadecyl (C18​H37​) alkyl chain, which confers high lipid solubility, and a polar, positively charged dimethyl-piperidinium phosphate head group. This duality is central to its ability to interact with and integrate into the phospholipid bilayers of cellular membranes. Laboratory-grade Perifosine is a solid with a purity exceeding 99% and demonstrates solubility in both aqueous and organic solvents, being soluble in water up to 10 mM and in ethanol up to 25 mM.[4]

Table 1: Drug Identification and Physicochemical Properties

ParameterValueSource(s)
Generic NamePerifosine1
DrugBank IDDB066411
CAS Number157716-52-41
TypeSmall Molecule1
Chemical FormulaC25​H52​NO4​P1
Average Molecular Weight461.668 g/mol1
IUPAC Name(1,1-Dimethylpiperidin-1-ium-4-yl) octadecyl hydrogen phosphate1
Key SynonymsKRX-0401, NSC-6399661

Chemical Classification and Analogs

Perifosine belongs to a distinct class of synthetic lipids whose therapeutic potential and challenges are intrinsically linked to their chemical structure.

  • Primary Classification: It is classified as a synthetic alkylphospholipid (APL) or, more specifically, an alkylphosphocholine (APC) analog.[4] This class of compounds is structurally related to naturally occurring lysophosphatidylcholine and includes other investigational agents such as miltefosine and edelfosine.[5]
  • Chemical Taxonomy: According to its functional groups, Perifosine is a dialkyl phosphate, defined as an organic compound containing a phosphate group esterified to two alkyl chains.[1] Its broader chemical hierarchy places it within the super class of organic acids and derivatives and the class of organic phosphoric acids and derivatives.[1]

The development of Perifosine was a direct evolution from earlier APLs like miltefosine. These first-generation compounds, while showing antineoplastic activity, were hampered in clinical development by severe, dose-limiting gastrointestinal (GI) toxicity.[11] This toxicity was attributed to the metabolic cleavage of the molecule, which released a phosphocholine metabolite structurally similar to the neurotransmitter acetylcholine, leading to potent parasympathomimetic effects like nausea, vomiting, and diarrhea.[11] The key structural innovation in Perifosine was the replacement of the labile choline head group with a stable heterocyclic piperidine moiety.[9] This modification was specifically designed to prevent the generation of the problematic phosphocholine metabolite and thereby improve the drug's therapeutic index.[11]

However, this rational design choice did not fully resolve the issue of GI toxicity. Across all of Perifosine's clinical trials, the most common and dose-limiting adverse events remained gastrointestinal in nature, including nausea, vomiting, and diarrhea.[7] While the specific acetylcholine-like mechanism was mitigated, the fundamental class property of being a membrane-inserting amphipathic molecule likely resulted in direct disruption of the cellular integrity of the GI tract lining. This suggests that a significant component of the observed GI toxicity is an intrinsic, on-target effect of this drug class, a factor that ultimately constrained dosing schedules and likely impacted patient tolerability and compliance during the prolonged treatment courses required in the pivotal Phase III trials.

Comprehensive Pharmacological Profile

Perifosine possesses a complex and multifaceted pharmacological profile. While primarily developed as an inhibitor of the Akt signaling pathway, its identity as a membrane-active alkylphospholipid confers additional, Akt-independent biological activities that contribute to its overall antineoplastic effects.

Mechanism of Action: Inhibition of the PI3K/Akt/mTOR Pathway

The central and most well-characterized mechanism of action for Perifosine is its unique mode of inhibiting the serine/threonine kinase Akt, a pivotal regulator of cellular survival and proliferation.

  • Unique Allosteric Inhibition: Most kinase inhibitors function by competing with adenosine triphosphate (ATP) for binding in the catalytic pocket of the target enzyme. Perifosine operates via a distinct, non-competitive mechanism. It is an allosteric inhibitor that does not interact with the ATP-binding site but instead targets the pleckstrin homology (PH) domain of Akt.[5]
  • Prevention of Membrane Translocation and Activation: The activation of Akt is a multi-step process initiated by its recruitment from the cytoplasm to the inner surface of the plasma membrane. This translocation is mediated by the binding of Akt's PH domain to the lipid second messengers phosphatidylinositol (3,4)-bisphosphate (PI(3,4)P2​) and phosphatidylinositol (3,4,5)-trisphosphate (PI(3,4,5)P3​), which are produced by phosphoinositide 3-kinase (PI3K).[10] Once localized at the membrane, Akt is phosphorylated at two key residues, threonine 308 (T308) by PDK1 and serine 473 (S473) by the mTORC2 complex, which is required for its full kinase activity.[10] By binding to or interfering with the function of the PH domain, Perifosine physically prevents Akt from translocating to the membrane, thereby blocking its access to its activating kinases.[10] Preclinical studies have consistently demonstrated that treatment with Perifosine leads to a dose-dependent reduction in the phosphorylation of Akt at Ser473, a key marker of its activation, without altering the total cellular levels of the Akt protein.[11]
  • Downstream Pathway Suppression: By preventing Akt activation, Perifosine effectively shuts down the entire downstream signaling cascade. This results in the reduced phosphorylation and altered activity of numerous Akt substrates that are critical for cell growth (e.g., mTOR, p70S6K), survival (e.g., BAD, XIAP), and metabolism (e.g., GSK3β), ultimately leading to cell cycle arrest and apoptosis.[10]

Mechanism of Action: Role as an Alkylphosphocholine (APC) Analog

Beyond its specific interaction with the Akt PH domain, Perifosine's chemical nature as a synthetic lipid endows it with broader membrane-centric activities that contribute to its anticancer effects.

  • Membrane Targeting and Disruption: As an amphipathic APL, Perifosine has a natural propensity to insert itself into phospholipid bilayers. This integration can alter the fundamental properties of cellular membranes, modulating their permeability, lipid composition, and overall function in signal transduction.[1]
  • Lipid Raft Interference: A critical aspect of this membrane activity is the disruption of specialized microdomains known as lipid rafts. These are regions of the plasma membrane enriched in cholesterol and sphingolipids that function as organizing centers or platforms for signaling complexes, including those required for the activation of receptor tyrosine kinases and Akt itself.[10] Malignant cells have been shown to possess a significantly higher density of these lipid rafts compared to normal cells, potentially offering a basis for the tumor-selective uptake and retention of APC analogs like Perifosine.[15] By accumulating in and disrupting the structure of these rafts, Perifosine can prevent the assembly of signaling machinery, providing a secondary, complementary mechanism for inhibiting Akt recruitment and activation.[10]
  • Modulation of Other Signaling Pathways: The cellular effects of Perifosine are not limited to the Akt pathway. Evidence indicates that it also inhibits the anti-apoptotic mitogen-activated protein kinase (MAPK) pathway and, crucially, activates the pro-apoptotic c-Jun N-terminal kinase (JNK) pathway.[1] In multiple myeloma cells, the activation of JNK has been described as an essential event for Perifosine-induced apoptosis, operating in parallel to or downstream of Akt inhibition.[16]
  • Upstream Effects: Research in malignant pleural mesothelioma cells has suggested that Perifosine can also exert effects upstream of Akt. It was shown to interfere with the phosphorylation and activation of receptor tyrosine kinases such as the epidermal growth factor receptor (EGFR) and MET, thereby reducing the initial signal that would normally lead to PI3K and Akt activation.[14]

This complex pharmacology, while appearing advantageous in a preclinical setting, presented a significant challenge for clinical development. The drug's pleiotropic effects, ranging from direct PH domain interaction to broad membrane disruption and modulation of multiple kinase pathways, made it difficult to pinpoint the single most critical mechanism of action driving its efficacy in any given tumor type. This ambiguity precluded the development of a specific, validated predictive biomarker. Without a reliable method to identify which patients' tumors were most vulnerable to Perifosine's specific mode of action—be it Akt-dependence, JNK activation, or lipid raft density—the pivotal Phase III trials had to enroll broad, unselected patient populations. This lack of a biomarker-driven patient selection strategy is a well-established factor contributing to the failure of targeted therapies in late-stage development, as any true efficacy signal in a small, responsive subset is likely to be diluted to non-significance within the larger study population.

Identified Molecular Targets

Based on its known mechanisms, Perifosine has been shown to interact with or modulate the activity of several key proteins in oncogenic signaling pathways.

  • Primary Target: RAC-alpha serine/threonine-protein kinase (Akt), with activity demonstrated against isoforms AKT1 and AKT3.[1]
  • Other Modulated Proteins:
  • Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform (PIK3CG) [1]
  • Mitogen-activated protein kinase 1 (MAPK1) [1]
  • Protein kinase C alpha type (PRKCA) [1]

Preclinical Evidence and Rationale for Clinical Development

The advancement of Perifosine into human clinical trials was supported by a robust body of preclinical evidence demonstrating its potent and broad-spectrum antineoplastic activity in both laboratory and animal models.

In Vitro Antineoplastic Activity

Initial laboratory studies established Perifosine as a promising anti-cancer agent with multiple cellular effects.

  • Broad Antiproliferative Activity: Perifosine demonstrated the ability to inhibit the growth of a wide variety of human tumor cell lines in vitro. This activity was observed in models of melanoma, nervous system cancers, lung, prostate, colon, and breast cancer, with half-maximal inhibitory concentrations (IC50) typically falling in the low micromolar range of 0.6 to 8.9 μM.[4]
  • Induction of Apoptosis and Cell Cycle Arrest: Mechanistic investigations confirmed that Perifosine's antiproliferative effects were mediated through the induction of programmed cell death (apoptosis) and the halting of the cell division cycle.[14] In mesothelioma cells, treatment with Perifosine led to a significant and progressive accumulation of cells in the G2/M phase of the cell cycle.[14] Furthermore, it was shown to induce the expression of the critical cell cycle regulator p21WAF1/CIP1 in a manner that was independent of the tumor suppressor p53, suggesting its activity would not be limited to p53-wildtype tumors.[8]
  • Synergistic Effects with Other Agents: A crucial component of the preclinical rationale was the observation that Perifosine could work synergistically with existing anticancer therapies. In colorectal cancer cell lines, Perifosine was shown to enhance the cytotoxic effects of 5-fluorouracil (5-FU). This synergy was linked to the inhibition of NF-κB, a transcription factor that is often activated by chemotherapy and promotes cell survival and resistance.[11] Similar synergistic or additive effects were observed when Perifosine was combined with cisplatin [14] and the proteasome inhibitor bortezomib [20], providing a strong basis for pursuing combination therapy strategies in the clinic.

In Vivo Efficacy in Animal Models

The promising in vitro results were successfully translated into animal models, confirming Perifosine's activity in a more complex biological system.

  • Tumor Growth Inhibition: Perifosine demonstrated substantial anticancer activity in vivo when administered to mice bearing human tumor xenografts.[4] In a key study using a human plasmacytoma mouse model, which mimics multiple myeloma, oral administration of Perifosine led to significant antitumor activity. This effect was directly correlated with a pharmacodynamic marker of target engagement: the downregulation of phosphorylated Akt in the tumor cells harvested from the treated animals.[16]
  • Efficacy in Brain Metastasis Models: One of the most compelling preclinical findings was Perifosine's activity against brain tumors. In orthotopic mouse models using human cancer cell lines originally isolated from brain metastases (prostate cancer DU 145 and lung cancer NCI-H1915), orally administered Perifosine was shown to effectively cross the blood-brain barrier, distribute into the brain, and remain localized in the brain tissue for an extended period.[13] This favorable distribution translated into significant therapeutic efficacy, with Perifosine treatment prolonging the survival of the tumor-bearing mice and even leading to complete tumor regression in the NCI-H1915 model.[13] This provided a powerful rationale for its clinical investigation in patients with primary brain cancers like glioblastoma.
  • Radiosensitizing Properties: In addition to its synergy with chemotherapy, preclinical studies also revealed that Perifosine could act as a radiosensitizer, enhancing the tumor-killing effects of radiation therapy.[7] This finding supported the initiation of a clinical trial to evaluate the safety and feasibility of combining Perifosine with radiotherapy in patients with advanced solid tumors.[7]

Preclinical Pharmacokinetics (ADME)

Pharmacokinetic studies in animals defined the absorption, distribution, metabolism, and excretion (ADME) profile of Perifosine, providing essential information for designing the first-in-human clinical trials.

  • Absorption: Perifosine was confirmed to be orally bioavailable, a key feature for patient convenience and long-term administration.[8] In mice, a single oral dose of 40 mg/kg was sufficient to achieve plasma concentrations considered to be clinically relevant.[23]
  • Distribution: Following absorption, the drug distributed widely throughout the body. While concentrations in the heart and brain were relatively low, significant accumulation was observed in tumor tissues.[23] Interestingly, the degree of tumor uptake varied between different xenograft models and appeared to correlate with the sensitivity of the tumor to the drug's cytotoxic effects, a finding that hinted at the importance of tumor-specific factors in determining response.[23] High levels of the drug were also found in the gastrointestinal tract, presaging the GI toxicities that would later be observed in clinical trials.[23]
  • Metabolism: A key pharmacokinetic characteristic of Perifosine is its metabolic stability. Studies showed that the drug was not metabolized in vivo, circulating and distributing as the parent compound.[23]
  • Elimination: Consistent with its high stability and lipophilicity, Perifosine displayed very slow elimination from the body. In mice, it exhibited a long terminal half-life of 137 ± 20 hours.[23]

The preclinical pharmacokinetic profile, while appearing favorable on the surface, concealed a potential clinical liability. The combination of good oral bioavailability, high stability, and a very long half-life suggested that consistent, steady-state drug exposure could be easily achieved, which led directly to the clinical dosing strategy of using a high loading dose followed by a lower daily maintenance dose.[11] However, for a drug with notable dose-dependent toxicities, a long half-life can be problematic. It means that if a patient experiences a severe adverse event, the drug takes a very long time to clear from their system, prolonging the toxicity and complicating its management. This slow clearance makes simple drug holidays less effective and can necessitate dose reductions or treatment discontinuations, which in turn may compromise therapeutic efficacy. The preclinical finding of high drug accumulation in the GI tract, coupled with this slow elimination, directly foreshadowed the clinical challenge of managing the GI side effects that became the drug's dose-limiting toxicity.

Clinical Development and Human Pharmacokinetics

The transition of Perifosine from preclinical models to human testing involved a series of Phase I studies to establish its safety and pharmacokinetic profile, which then guided the design of later-phase efficacy trials.

Phase I Clinical Trials and Safety Profile

Initial human studies were designed to determine the maximum tolerated dose (MTD), define the dose-limiting toxicities (DLTs), and establish a recommended Phase II dose (RP2D) and schedule.

  • Dose Escalation and Dosing Schedule: Perifosine was evaluated in Phase I trials across various dosing schedules, including daily, weekly, and daily administration following an initial loading dose.[25] A pivotal Phase I study in patients with refractory neoplasms utilized a loading dose/maintenance dose schedule. This trial established an MTD and RP2D consisting of a 900 mg loading dose on day 1 (divided into two doses), followed by a maintenance dose of 150 mg daily.[24] A separate Phase I trial in pediatric patients with recurrent solid tumors and CNS tumors established a pediatric RP2D of 50 mg/m²/day.[26]
  • Dose-Limiting Toxicities (DLTs): Across these early trials, the DLTs were consistently and predominantly gastrointestinal in nature. In the loading dose study, dose-limiting diarrhea was observed at dose levels of 1,200 mg (loading) and 200 mg (maintenance) or higher.[24] In a Phase I trial combining Perifosine with concurrent radiation therapy, DLTs of nausea and vomiting occurred at a daily dose of 200 mg.[7]
  • Early Efficacy Signals: Although the primary objective of Phase I trials is to assess safety, they often provide preliminary indications of antitumor activity. In the case of Perifosine, single-agent activity was observed, with several patients experiencing prolonged periods of stable disease, including in sarcoma and other refractory neoplasms.[24]

Human Pharmacokinetic Profile

Pharmacokinetic analyses from the Phase I trials largely confirmed the predictions from preclinical models, defining how the drug is absorbed, distributed, and eliminated in humans. A summary of key parameters is presented in Table 2.

  • Dose-Dependent Exposure and Long Half-Life: The studies confirmed that plasma concentrations of Perifosine were dose-dependent. Due to its slow elimination, steady-state plasma levels were typically reached after approximately one week of continuous daily dosing.[7] The terminal half-life in humans was confirmed to be very long, calculated at 105 hours in one analysis, and drug accumulation was observed across multiple treatment cycles.[11]
  • Achievement of Therapeutic Concentrations: A critical finding from the Phase I studies was that the established MTD resulted in drug concentrations that were within the biologically active range identified in in vitro experiments. At the RP2D of 900 mg loading/150 mg maintenance, the median peak plasma concentration (Cmax​) reached was approximately 8.3 µg/mL.[24] This concentration, equivalent to approximately 18 µM, comfortably exceeds the IC50 values (0.6-8.9 µM) observed in preclinical cell culture assays, providing confidence that the clinical dosing was sufficient to achieve a biological effect.[4]

Table 2: Summary of Key Preclinical and Clinical Pharmacokinetic Parameters

ParameterPreclinical Finding (Mouse Model)Clinical Finding (Human)Source(s)
BioavailabilityOrally bioavailableOrally bioavailable13
MetabolismNot metabolizedNot metabolized23
Key Distribution SitesWhole body, high in GI tract and tumors, low in brain/heartNot fully detailed, but brain penetration suggested13
Terminal Half-Life (t1/2​)137 ± 20 hours~105 hours11
Time to Steady StateNot specified~1 week7
Clinically Achieved Cmax​Clinically relevant levels at 40 mg/kg~8.3 µg/mL (~18 µM) at MTD23

Consolidated Safety and Tolerability Profile

Data aggregated from across the entire clinical development program provide a consistent and clear picture of Perifosine's safety profile.

  • Most Common Adverse Events: The most frequently reported drug-related toxicities were consistently low-grade and manageable. They were dominated by gastrointestinal side effects, including nausea (50-74%), vomiting (38-61%), and diarrhea (32-42%), and constitutional symptoms such as fatigue (31-48%) and anorexia (19%).[7] Musculoskeletal pain was also reported in 20-46% of patients in some trials.[12]
  • Lack of Myelosuppression: A significant and favorable aspect of Perifosine's safety profile was the conspicuous absence of meaningful bone marrow toxicity. Unlike most cytotoxic chemotherapies, Perifosine did not cause significant myelosuppression, neutropenia, or thrombocytopenia when used as a single agent.[7] This characteristic made it an ideal candidate for combination with myelosuppressive agents, as it was not expected to exacerbate their hematologic toxicities.
  • Overall Tolerability: Despite the high frequency of GI adverse events, Perifosine was generally described in clinical trial reports as well-tolerated, with most toxicities being Grade 1 or 2 and manageable with standard supportive care (e.g., antiemetics, antidiarrheals) and dose modifications.[12] However, it is noteworthy that in some Phase II studies, dose reductions due to adverse events were required in a substantial minority of patients, around 17-20%.[12] This indicates that while not life-threatening, the cumulative burden of low-grade toxicities could impact a patient's ability to remain on the full dose of the drug for extended periods.

Critical Analysis of Clinical Trials by Indication

Perifosine was investigated across a wide range of malignancies, but its development was most advanced in multiple myeloma and metastatic colorectal cancer, where it progressed to pivotal Phase III trials. The outcomes of these trials, contrasted with their promising predecessors, are central to understanding the drug's ultimate failure.

Multiple Myeloma (MM)

The PI3K/Akt pathway is a well-validated therapeutic target in multiple myeloma, making it a logical indication for Perifosine.

  • Early Promise (Phase I/II): Initial clinical studies of Perifosine in combination with the proteasome inhibitor bortezomib, with or without dexamethasone, yielded highly encouraging results in heavily pre-treated patients with relapsed/refractory MM. An updated analysis of a Phase I/II study involving 73 evaluable patients demonstrated an overall response rate (ORR, defined as partial response or better) of 38%.[31] The median time to progression (TTP) was 6.4 months, and the median overall survival (OS) was an impressive 22.5 months. The activity was particularly notable in patients who had previously relapsed after bortezomib therapy (ORR 55%) and even in those refractory to bortezomib (ORR 32%, median OS 16 months).[31] These data suggested that Perifosine could overcome or re-sensitize tumors to proteasome inhibition and provided a robust rationale for proceeding to a confirmatory Phase III trial.
  • Pivotal Failure (Phase III - NCT01002248): Based on the promising Phase I/II data, a large, international, randomized, double-blind, placebo-controlled Phase III study was initiated. The trial was designed to evaluate the efficacy of adding Perifosine (50 mg daily) to a standard regimen of bortezomib and dexamethasone in patients with MM who had relapsed after at least one prior bortezomib-containing therapy.[22] The primary endpoint was progression-free survival (PFS).[22] In March 2013, it was announced that the trial was being discontinued. The decision was based on the recommendation of an independent data safety and monitoring committee following a pre-planned interim analysis, which concluded that the study was highly unlikely to meet its primary endpoint.[5]
  • Efficacy Results: The interim analysis revealed that the addition of Perifosine not only failed to provide a benefit but trended toward a worse outcome for the primary endpoint. The median PFS was 22.7 weeks in the Perifosine arm compared to 39.0 weeks in the placebo arm, corresponding to a hazard ratio (HR) of 1.269 (95% CI: 0.817, 1.969; p=0.287).[22] The overall response rate was also lower in the Perifosine arm (20.3%) versus the placebo arm (27.3%).[22]
  • A Paradoxical Survival Signal: A perplexing finding from the interim analysis was a contradictory trend in overall survival. Despite the negative result for PFS, the median OS was numerically longer in the Perifosine arm (141.9 weeks) than in the placebo arm (83.3 weeks), with an HR of 0.734 (95% CI: 0.380, 1.419; p=0.356).[22] While not statistically significant and based on a small number of events due to the early termination of the trial, this discordance between PFS and OS is highly unusual. It raises complex questions about the drug's biological effects. It is possible that Perifosine, while not delaying initial progression, may have had a longer-term biological effect not captured by standard response criteria, or that differences in subsequent therapies confounded the OS analysis. However, given the immaturity of the data, this finding could also represent a statistical anomaly.
  • Safety: The trial did not raise any new safety concerns. The incidence of Grade 3/4 adverse events was comparable between the arms (61% for Perifosine vs. 55% for placebo), though thrombocytopenia (26% vs. 14%) and pneumonia (9% vs. 3%) were more frequent in the Perifosine group.[22]

Metastatic Colorectal Cancer (mCRC)

The development program for Perifosine in mCRC stands as a textbook example of the "Phase II to Phase III disconnect," where highly promising early results fail to be validated in a larger confirmatory trial.

  • Phase II Success (NCT00398879): A multicenter, randomized, double-blind, placebo-controlled Phase II trial evaluated Perifosine plus capecitabine (P-CAP) against capecitabine plus placebo (CAP) in 38 patients with mCRC who had progressed on prior therapies.[38] The results were exceptionally positive.
  • Efficacy Results: The addition of Perifosine to capecitabine resulted in a dramatic and highly statistically significant improvement in efficacy. The median time to progression (TTP) was more than doubled, from 10.1 weeks in the CAP arm to 27.5 weeks in the P-CAP arm (p < 0.001).[38] Similarly, median overall survival was significantly extended from 7.6 months to 17.7 months (p = 0.0052).[38] The ORR was also higher in the combination arm (20% vs. 7%).[38] Based on these compelling results, the FDA granted Fast Track designation for Perifosine in this indication.[41]
  • Pivotal Failure (Phase III - X-PECT Study): Based on the strength of the Phase II data, a large (n=468), similarly designed, randomized, double-blind, placebo-controlled Phase III trial was initiated.[43] The primary endpoint was overall survival. The results of the X-PECT study, announced in April 2012, were unequivocally negative.[5] The study showed no benefit in overall survival for adding Perifosine to capecitabine in the refractory mCRC setting.[44] Secondary endpoints, including progression-free survival and response rate, also showed no improvement with the addition of Perifosine.[44]

The stark contrast between the Phase II and Phase III results in mCRC highlights the inherent risks of drug development. The positive Phase II finding, generated in a very small study population (n=38), was likely a statistical anomaly (a Type I error), where random chance may have led to an imbalance of patients with favorable prognostic factors in the experimental arm. When the same therapeutic question was tested in a much larger, more statistically robust population, the true null effect was revealed. This outcome underscores the critical importance of large, well-powered, confirmatory Phase III trials and serves as a powerful cautionary tale against over-interpreting dramatic results from small, early-phase studies.

Malignant Glioma (Glioblastoma - GBM)

Given the strong preclinical data showing Perifosine's ability to penetrate the central nervous system and its efficacy in brain tumor models, glioblastoma was a rational target for clinical investigation.

  • Monotherapy Inefficacy (Phase II - NCT00590954): A Phase II, single-arm trial was conducted to evaluate Perifosine monotherapy in patients with recurrent GBM.[45] The trial enrolled 16 patients with GBM and treated them with a 600 mg loading dose followed by 100 mg daily. The results were disappointing. The primary endpoint, 6-month progression-free survival (PFS6), was 0%.[45] The median PFS was a mere 1.58 months, and the median OS was 3.68 months, with no radiographic responses observed. The study concluded that Perifosine is ineffective as a single agent for recurrent GBM.[45]
  • Combination Approaches (Phase I/II - NCT01051557): Recognizing the limitations of monotherapy in this aggressive disease, investigators initiated a Phase I trial to evaluate the combination of Perifosine with the mTOR inhibitor temsirolimus, aiming for a dual blockade of the PI3K/Akt/mTOR pathway.[47] The study enrolled 35 patients with recurrent malignant gliomas and successfully established that the combination was tolerable. However, the development of Perifosine was halted before this combination strategy could be tested for efficacy in a Phase II component.[47]

Investigations in Other Malignancies

The clinical development program for Perifosine was broad, with studies conducted in several other tumor types, generally with modest results.

  • Renal Cell Carcinoma (RCC): Phase II trials of Perifosine in patients with advanced RCC following failure of tyrosine kinase inhibitors (TKIs) or mTOR inhibitors showed evidence of clinical activity. In a combined analysis of two studies, the ORR was 8%, and 36% of patients achieved stable disease, suggesting a modest single-agent benefit in this setting.[12]
  • Non-Small Cell Lung Cancer (NSCLC): A completed clinical trial in NSCLC is documented, but specific efficacy results are not available in the provided materials.[50]
  • Pediatric Solid Tumors: A Phase I dose-escalation study (NCT00776867) evaluated single-agent Perifosine in 23 children with recurrent or refractory solid tumors, including neuroblastoma and high-grade gliomas.[26] The drug was found to be safe and feasible in this population. While no objective responses were observed, five patients with neuroblastoma achieved stable disease, which was prolonged (≥11 months) in three of them.[26]

Table 3: Comparative Efficacy Outcomes of Pivotal Perifosine Clinical Trials

IndicationTrial PhasePrimary EndpointPerifosine Arm ResultControl Arm ResultHazard Ratio (95% CI)p-ValueSource(s)
mCRCPhase IITTP27.5 weeks10.1 weeksNot Reported< 0.00138
mCRCPhase III (X-PECT)OSNo benefitNo benefitNot ReportedNot Significant44
MMPhase IIIPFS22.7 weeks39.0 weeks1.269 (0.817, 1.969)0.28722

Regulatory History and Final Development Status

The regulatory pathway of Perifosine reflects its initial clinical promise and subsequent definitive failure.

  • Fast Track Designation: In recognition of its potential to address significant unmet medical needs in life-threatening diseases, the U.S. Food and Drug Administration (FDA) granted Fast Track designation to Perifosine for two separate indications: relapsed/refractory multiple myeloma in December 2009 [52] and refractory advanced colorectal cancer in April 2010.[41]
  • Orphan Drug Designation: Perifosine also received Orphan Drug designation, a status granted to drugs intended for rare diseases. In 2010, it was designated for multiple myeloma and neuroblastoma in the U.S., and for multiple myeloma by the European Medicines Agency (EMA).[5]
  • Withdrawal and Discontinuation: The clinical development program for Perifosine was terminated following the negative outcomes of the pivotal Phase III trials. The failure of the X-PECT study in mCRC was announced in April 2012, and the discontinuation of the MM trial was announced in March 2013.[5] Following the cessation of its development, the previously granted Orphan Drug designations were formally withdrawn by the sponsors, with the neuroblastoma designation being revoked in March 2017.[5]
  • Current Status: Perifosine is not an approved medication in any jurisdiction and is considered a discontinued investigational drug candidate.[2] Its development was managed by Keryx Biopharmaceuticals, who had licensed the compound from Æterna Zentaris Inc.[5] Following the trial failures, the licensing agreement was terminated, and the rights reverted to Æterna Zentaris.[55]

Synthesis and Expert Conclusion

The clinical development of Perifosine offers a compelling and instructive narrative on the complexities and risks inherent in oncology drug discovery. Despite a strong preclinical rationale, a novel mechanism targeting a critical oncogenic pathway, and highly encouraging Phase II data, the drug ultimately failed to demonstrate a clinical benefit in large, confirmatory Phase III trials. The downfall of Perifosine can be attributed to a confluence of factors that serve as critical lessons for the field.

First and foremost, the Perifosine story highlights the biomarker imperative. The PI3K/Akt pathway is a central signaling hub that is dysregulated in a majority of human cancers, but its ubiquitous nature is a double-edged sword. Targeting such a fundamental pathway without a validated predictive biomarker to identify the subset of patients whose tumors are truly "addicted" to Akt signaling is a high-risk strategy. The broad, unselected patient populations enrolled in the Phase III trials for mCRC and MM likely contained only a small, unidentified fraction of true responders, and their potential benefit was completely diluted by the lack of effect in the majority. The failure to develop and integrate a robust biomarker strategy was arguably the single greatest strategic flaw in the drug's development.

Second, the complex, pleiotropic mechanism of action, while a source of potent activity in preclinical models, became a liability in the clinical setting. As both a specific allosteric Akt inhibitor and a general membrane-disrupting agent that also modulated the JNK and MAPK pathways, it was never entirely clear which of its activities was the primary driver of efficacy. This mechanistic ambiguity further complicated any potential biomarker strategy and made it difficult to rationally design combination therapies or select appropriate patient populations.

Third, the mCRC program is a stark reminder of the Phase II-to-III chasm. The dramatic, statistically significant survival benefit seen in the small, 38-patient Phase II trial created high expectations but ultimately proved to be a statistical aberration that did not hold up under the rigor of a properly powered, 468-patient Phase III study. This outcome serves as a powerful cautionary tale about the dangers of over-interpreting results from small, early-phase trials and underscores their role as hypothesis-generating, not definitive, evidence.

Finally, the drug's pharmacokinetic profile presented subtle but significant clinical challenges. The very long half-life, initially viewed as favorable for maintaining drug exposure, likely narrowed the therapeutic window by making the management of the predictable, on-target GI toxicities more difficult. The inability to quickly clear the drug during periods of toxicity may have led to more frequent dose interruptions and reductions, potentially compromising the sustained therapeutic pressure needed for efficacy in the long-term Phase III setting.

In conclusion, Perifosine, a molecule of significant scientific interest, failed not because of a lack of biological activity or an unacceptable safety profile, but because of an inability to translate its broad preclinical effects into a demonstrable clinical benefit in a well-defined patient population. Its history reinforces the modern paradigm of oncology drug development, which emphasizes a deep understanding of mechanism, the co-development of predictive biomarkers, and a cautious interpretation of early-phase clinical data. Perifosine remains a critical case study for pharmacologists, clinical researchers, and drug developers, illustrating the immense challenges of drugging the Akt pathway and the essential role of precision medicine in realizing the potential of targeted therapies.

Works cited

  1. Perifosine: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed August 25, 2025, https://go.drugbank.com/drugs/DB06641
  2. Disclaimer: This resource is intended for purely research purposes. It should not be used for emergencies or medical or professional advice. - DGIdb, accessed August 25, 2025, https://dgidb.org/drugs/ncit:C1727
  3. Perifosine(KRX-0401) , 10mMinWater , 157716-52-4 - CookeChem, accessed August 25, 2025, https://www.cookechem.com/Detail/A7701358.htm
  4. Perifosine, Akt inhibitor (CAS 157716-52-4) | Abcam, accessed August 25, 2025, https://www.abcam.com/en-us/products/biochemicals/perifosine-akt-inhibitor-ab142055
  5. Perifosine - Wikipedia, accessed August 25, 2025, https://en.wikipedia.org/wiki/Perifosine
  6. CAS 157716-52-4 | Perifosine (KRX-0401) supply - Guidechem, accessed August 25, 2025, https://www.guidechem.com/supply/157716-52-4.html
  7. Phase I and pharmacokinetic study of combined treatment with ..., accessed August 25, 2025, https://pubmed.ncbi.nlm.nih.gov/16914220/
  8. Perifosine, a novel alkylphospholipid, inhibits protein kinase B activation - AACR Journals, accessed August 25, 2025, https://aacrjournals.org/mct/article/2/11/1093/234119/Perifosine-a-novel-alkylphospholipid-inhibits
  9. A Phase I Trial of Perifosine (NSC 639966) on a Loading Dose/Maintenance Dose Schedule in Patients with Advanced Cancer - AACR Journals, accessed August 25, 2025, https://aacrjournals.org/clincancerres/article/10/22/7450/185350/A-Phase-I-Trial-of-Perifosine-NSC-639966-on-a
  10. Perifosine: Update on a Novel Akt Inhibitor - PMC, accessed August 25, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6957247/
  11. Perifosine, an oral, anti-cancer agent and inhibitor of the Akt pathway: mechanistic actions, pharmacodynamics, pharmacokinetics, and clinical activity - PMC, accessed August 25, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4467022/
  12. Two Phase II Trials of the Novel Akt Inhibitor Perifosine in Patients with Advanced Renal Cell Carcinoma Following Progression on VEGF-Targeted Therapy - PMC, accessed August 25, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3934426/
  13. Perifosine, a Bioavailable Alkylphospholipid Akt Inhibitor, Exhibits Antitumor Activity in Murine Models of Cancer Brain Metastasis Through Favorable Tumor Exposure - Frontiers, accessed August 25, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2021.754365/full
  14. Perifosine as a Potential Novel Anti-Cancer Agent Inhibits EGFR/MET-AKT Axis in Malignant Pleural Mesothelioma - Semantic Scholar, accessed August 25, 2025, https://pdfs.semanticscholar.org/03cc/2ed77a3d7808bdfed4aa5bda7c5ff5900cd8.pdf
  15. Clinical Imaging and Dosimetry of a Pan-Cancer Targeting Alkylphosphocholine Analog, [ 124 I]I-NM404 - MDPI, accessed August 25, 2025, https://www.mdpi.com/2673-592X/2/2/15
  16. Perifosine, an oral bioactive novel alkylphospholipid, inhibits Akt and induces in vitro and in vivo cytotoxicity in human multiple myeloma cells | Blood, accessed August 25, 2025, https://ashpublications.org/blood/article/107/10/4053/109834/Perifosine-an-oral-bioactive-novel
  17. Protein Annotation DataBase - Search - ASpdb, accessed August 25, 2025, https://biodataai.uth.edu/ASpdb/gene_search_result.cgi?page=page&type=quick_search&quick_search=5594
  18. Perifosine (KRX-0401) | Akt Inhibitor | MedChemExpress, accessed August 25, 2025, https://www.medchemexpress.com/Perifosine.html
  19. Perifosine | C25H52NO4P | CID 148177 - PubChem, accessed August 25, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Perifosine
  20. Perifosine, an oral, anti-cancer agent and inhibitor of the Akt pathway: Mechanistic actions, pharmacodynamics, pharmacokinetics, and clinical activity | Request PDF - ResearchGate, accessed August 25, 2025, https://www.researchgate.net/publication/224707341_Perifosine_an_oral_anti-cancer_agent_and_inhibitor_of_the_Akt_pathway_Mechanistic_actions_pharmacodynamics_pharmacokinetics_and_clinical_activity
  21. Perifosine , an oral, anti-cancer agent and inhibitor of the Akt pathway: mechanistic actions, pharmacodynamics, pharmacokinetics, and clinical activity - PubMed, accessed August 25, 2025, https://pubmed.ncbi.nlm.nih.gov/22512706/
  22. Randomized, placebo‐controlled, phase 3 study of perifosine combined with bortezomib and dexamethasone in patients with relapsed, refractory multiple myeloma previously treated with bortezomib - PMC - PubMed Central, accessed August 25, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9175725/
  23. Tumor and normal tissue pharmacokinetics of perifosine, an oral anti ..., accessed August 25, 2025, https://pubmed.ncbi.nlm.nih.gov/16012787/
  24. A phase I and pharmacokinetic study of oral perifosine with different ..., accessed August 25, 2025, https://pubmed.ncbi.nlm.nih.gov/25183650/
  25. Open Label Trial of Perifosine in Patients Currently Being Treated on Perifosine Trials in Solid Tumors or Multiple Myeloma | ClinicalTrials.gov, accessed August 25, 2025, https://clinicaltrials.gov/study/NCT00847366
  26. A phase I study of single-agent perifosine for recurrent or refractory pediatric CNS and solid tumors | PLOS One - Research journals, accessed August 25, 2025, https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0178593
  27. Perifosine: Update on a novel Akt inhibitor - Johns Hopkins University, accessed August 25, 2025, https://pure.johnshopkins.edu/en/publications/perifosine-update-on-a-novel-akt-inhibitor-4
  28. Phase I study of perifosine monotherapy in patients with recurrent or refractory neuroblastoma - PubMed, accessed August 25, 2025, https://pubmed.ncbi.nlm.nih.gov/28521076/
  29. Perifosine (KRX-0401) - The Myeloma Beacon, accessed August 25, 2025, https://myelomabeacon.org/resources/2009/11/04/perifosine/
  30. Two Phase II Trials of the Novel Akt Inhibitor Perifosine in Patients ..., accessed August 25, 2025, https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3934426/
  31. Perifosine in Combination with Bortezomib and Dexamethasone Extends Progression-Free Survival and Overall Survival in Relapsed/Refractory Multiple Myeloma Patients Previously Treated with Bortezombib: Updated Phase I/II Trial Results. | Blood | American Society of Hematology, accessed August 25, 2025, https://ashpublications.org/blood/article/114/22/1869/111262/Perifosine-in-Combination-with-Bortezomib-and
  32. Phase II Trial of Perifosine (KRX-0401) in Relapsed and/or Refractory Waldenström Macroglobulinemia: Preliminary Results. | Blood | American Society of Hematology, accessed August 25, 2025, https://ashpublications.org/blood/article/110/11/4493/117779/Phase-II-Trial-of-Perifosine-KRX-0401-in-Relapsed
  33. Perifosine--a new option in treatment of acute myeloid leukemia? - PubMed, accessed August 25, 2025, https://pubmed.ncbi.nlm.nih.gov/23931614/
  34. Randomized Placebo-Controlled Phase III Study Of Perifosine ..., accessed August 25, 2025, https://ashpublications.org/blood/article/122/21/3189/476269/Randomized-Placebo-Controlled-Phase-III-Study-Of
  35. Assessment of Efficacy and Safety of Perifosine, Bortezomib and Dexamethasone in Multiple Myeloma Patients | ClinicalTrials.gov, accessed August 25, 2025, https://clinicaltrials.gov/study/NCT01002248
  36. Aeterna Zentaris halts late-stage study of perifosine in multiple myeloma - FirstWord Pharma, accessed August 25, 2025, https://firstwordpharma.com/story/1638595
  37. Aeterna Zentaris to Discontinue Phase 3 Trial in Multiple Myeloma with Perifosine Following Data Safety Monitoring Board Recommendation | Fierce Biotech, accessed August 25, 2025, https://www.fiercebiotech.com/biotech/aeterna-zentaris-to-discontinue-phase-3-trial-multiple-myeloma-perifosine-following-data
  38. Randomized placebo-controlled phase II trial of perifosine plus ..., accessed August 25, 2025, https://pubmed.ncbi.nlm.nih.gov/21969495/
  39. Placebo-Controlled Study of Perifosine + Single Agent Chemotherapy for Metastatic Cancer Patients | ClinicalTrials.gov, accessed August 25, 2025, https://clinicaltrials.gov/study/NCT00398879
  40. Randomized Placebo-Controlled Phase II Trial of Perifosine Plus Capecitabine As Second- or Third-Line Therapy in Patients With Metastatic Colorectal Cancer - ASCO Publications, accessed August 25, 2025, https://ascopubs.org/doi/10.1200/JCO.2011.36.1980
  41. Keryx Receives FDA Fast Track Designation for KRX-0401 (Perifosine) for the Treatment of Refractory Advanced Colorectal Cancer | Fierce Biotech, accessed August 25, 2025, https://www.fiercebiotech.com/biotech/keryx-receives-fda-fast-track-designation-for-krx-0401-perifosine-for-treatment-of
  42. FDA Approves Fast Track Status for Perifosine in Colorectal Cancer - Medscape, accessed August 25, 2025, https://www.medscape.com/viewarticle/719787
  43. X-PECT Study of Perifosine (KRX-0401) for the Treatment of Refractory Advanced Colorectal Cancer to Proceed - Onco'Zine, accessed August 25, 2025, https://www.oncozine.com/x-pect-study-of-perifosine-krx-0401-for-the-treatment-of-refractory-advanced-colorectal-cancer-to-proceed/
  44. Results of the X-PECT study: A phase III randomized double-blind, placebo-controlled study of perifosine plus capecitabine (P-CAP) versus placebo plus capecitabine (CAP) in patients (pts) with refractory metastatic colorectal cancer (mCRC). | Journal of Clinical Oncology - ASCO Publications, accessed August 25, 2025, https://ascopubs.org/doi/10.1200/jco.2012.30.18_suppl.lba3501
  45. Phase II trial of an AKT inhibitor (perifosine) for recurrent ..., accessed August 25, 2025, https://www.researchgate.net/publication/334575448_Phase_II_trial_of_an_AKT_inhibitor_perifosine_for_recurrent_glioblastoma
  46. Study Results | Clinical and Molecular-Metabolic Phase II Trial of Perifosine for Recurrent/Progressive Malignant Gliomas | ClinicalTrials.gov, accessed August 25, 2025, https://www.clinicaltrials.gov/study/NCT00590954?term=PERIFOSINE&viewType=Table&rank=7&tab=results
  47. Phase I clinical trial of temsirolimus and perifosine for recurrent glioblastoma - PubMed, accessed August 25, 2025, https://pubmed.ncbi.nlm.nih.gov/32293798/
  48. Temsirolimus and Perifosine in Treating Patients With Recurrent or Progressive Malignant Glioma | ClinicalTrials.gov, accessed August 25, 2025, https://clinicaltrials.gov/study/NCT01051557
  49. Adenocarcinoma, Renal Completed Phase 2 Trials for ... - DrugBank, accessed August 25, 2025, https://go.drugbank.com/indications/DBCOND0126095/clinical_trials/DB06641?phase=2&status=completed
  50. Non-Small Cell Lung Cancer Completed Phase Trials for Perifosine (DB06641) - DrugBank, accessed August 25, 2025, https://go.drugbank.com/indications/DBCOND0132865/clinical_trials/DB06641?phase=&status=completed
  51. Perifosine Completed Phase 1 Trials for Solid Tumors Treatment | DrugBank Online, accessed August 25, 2025, https://go.drugbank.com/drugs/DB06641/clinical_trials?conditions=DBCOND0029860&phase=1&purpose=treatment&status=completed
  52. Keryx Receives FDA Fast Track Designation for KRX-0401 (Perifosine) for the Treatment of Relapsed/Refractory Multiple Myeloma | Fierce Biotech, accessed August 25, 2025, https://www.fiercebiotech.com/biotech/keryx-receives-fda-fast-track-designation-for-krx-0401-perifosine-for-treatment-of-relapsed
  53. Search Orphan Drug Designations and Approvals - FDA, accessed August 25, 2025, https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=310010
  54. Study Details | Perifosine and Torisel (Temsirolimus) for Recurrent/Progressive Malignant Gliomas | ClinicalTrials.gov, accessed August 25, 2025, https://clinicaltrials.gov/study/NCT02238496?term=AREA%5BBasicSearch%5D(AREA%5BConditionSearch%5D(%22Anaplastic%20Astrocytoma%22)%20AND%20AREA%5BInterventionSearch%5D(%22Temsirolimus%22))&rank=2
  55. AEterna Zentaris, Keryx Biopharmaceuticals End Perifosine License Pact; Aeterna to Continue Trials - BioSpace, accessed August 25, 2025, https://www.biospace.com/aeterna-zentaris-keryx-biopharmaceuticals-end-perifosine-license-pact-b-aeterna-b-to-continue-trials

Published at: August 25, 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.