Small Molecule
C32H41N9O4
1197160-78-3
Gedatolisib (PF-05212384, PKI-587) is an investigational, intravenously administered, small molecule therapeutic poised to become a first-in-class dual inhibitor of the phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin (mTOR) signaling pathways. Developed by Celcuity Inc., this agent is distinguished by its unique mechanism of action, which involves the potent, comprehensive, and simultaneous inhibition of all four Class I PI3K isoforms (p110α, β, γ, δ) and both critical mTOR complexes (mTORC1 and mTORC2). This complete blockade of the PI3K/AKT/mTOR (PAM) signaling cascade is designed to overcome the adaptive resistance mechanisms that have limited the efficacy of previous single-node pathway inhibitors.
The clinical development of Gedatolisib is anchored by the pivotal Phase 3 VIKTORIA-1 trial, which has yielded practice-changing results in a population with a profound unmet medical need: patients with hormone receptor-positive (HR+), HER2-negative advanced breast cancer (ABC) whose disease has progressed following treatment with a CDK4/6 inhibitor. In the PIK3CA wild-type cohort, a population with historically poor outcomes, Gedatolisib in combination with palbociclib and fulvestrant demonstrated an unprecedented 76% reduction in the risk of disease progression or death, extending median progression-free survival (PFS) to 9.3 months compared to 2.0 months with fulvestrant alone.
This remarkable efficacy is complemented by a highly favorable safety and tolerability profile. Notably, Gedatolisib is associated with low rates of severe hyperglycemia and stomatitis, common dose-limiting toxicities for this class of agents. The treatment discontinuation rate due to adverse events in the VIKTORIA-1 trial was exceptionally low (<4%), a key differentiator that enables sustained therapy and maximization of clinical benefit.
Recognizing its transformative potential, the U.S. Food and Drug Administration (FDA) has granted Gedatolisib Fast Track and Breakthrough Therapy designations and has accepted its New Drug Application (NDA) for review under the Real-Time Oncology Review (RTOR) program, signaling an accelerated path to potential approval. With promising early data also emerging in other malignancies, including metastatic castration-resistant prostate cancer (mCRPC), Gedatolisib represents a significant therapeutic advance, with the potential to establish a new standard of care and fundamentally alter the treatment landscape for a broad spectrum of solid tumors driven by PAM pathway dysregulation.
The therapeutic potential of Gedatolisib is rooted in its distinct chemical structure and a precisely engineered mechanism of action designed for comprehensive and durable inhibition of one of cancer's most critical signaling networks. A thorough examination of its pharmacological properties provides the foundational rationale for its clinical development and observed efficacy.
Gedatolisib is a synthetic organic compound classified as a small molecule therapeutic.[1] Its definitive chemical formula is
C32H41N9O4, corresponding to an average molecular weight of approximately 615.74 g/mol and a monoisotopic mass of 615.3282 g/mol.[2] It is identified globally by its Chemical Abstracts Service (CAS) Registry Number, 1197160-78-3, and is cataloged in the DrugBank database under the accession number DB11896.[1]
During its development, the compound has been referred to by several synonyms and code names, most notably the Pfizer designation PF-05212384 and the earlier identifier PKI-587.[1] The formal International Union of Pure and Applied Chemistry (IUPAC) name for the molecule is 1-phenyl]-3-[4-(4,6-dimorpholin-4-yl-1,3,5-triazin-2-yl)phenyl]urea.[5] Chemically, it belongs to the class of 1-benzoylpiperidines, characterized by a piperidine ring substituted at the 1-position with a benzoyl group.[2]
Physically, Gedatolisib presents as an off-white solid.[7] Its solubility profile is a critical determinant of its formulation and administration route; it is characterized as being insoluble in water and ethanol but is soluble in dimethyl sulfoxide (DMSO), particularly with gentle warming.[7] An analysis of its molecular properties against Lipinski's Rule of Five, a guideline for predicting oral bioavailability, reveals two violations: a molecular weight greater than 500 Da and more than 10 hydrogen bond acceptors (13 total).[4] These characteristics are often associated with poor membrane permeability and low oral absorption. This physicochemical profile provides a strong scientific basis for the selection of an intravenous route of administration for Gedatolisib.[1] Intravenous delivery bypasses the gastrointestinal tract, ensuring 100% bioavailability and leading to more predictable and consistent pharmacokinetic profiles, which is particularly advantageous for a potent kinase inhibitor with a narrow therapeutic window.
| Property | Value | Source(s) |
|---|---|---|
| Generic Name | Gedatolisib | 1 |
| DrugBank ID | DB11896 | 2 |
| Type | Small Molecule | 1 |
| Developer | Celcuity Inc. (licensed from Pfizer) | 11 |
| Synonyms | PF-05212384, PKI-587 | 1 |
| CAS Number | 1197160-78-3 | 1 |
| Chemical Formula | C32H41N9O4 | 1 |
| Molecular Weight | 615.74 g/mol | 4 |
| IUPAC Name | 1-phenyl]-3-[4-(4,6-dimorpholin-4-yl-1,3,5-triazin-2-yl)phenyl]urea | 5 |
| Administration Route | Intravenous | 1 |
| Solubility | Insoluble in water; Soluble in DMSO | 7 |
Gedatolisib is a potent and reversible dual inhibitor that simultaneously targets two of the most critical regulatory nodes within the PI3K/AKT/mTOR (PAM) signaling pathway.[1] This pathway is a central command-and-control system for fundamental cellular processes, including growth, proliferation, survival, and metabolism. Its frequent dysregulation through genetic mutations or aberrant activation is a well-established driver of oncogenesis and therapeutic resistance across a multitude of human cancers.[5]
The inhibitory activity of Gedatolisib is characterized by its breadth and potency. It functions as a pan-Class I PI3K inhibitor, demonstrating robust, low-nanomolar activity against all four of the Class I catalytic subunit isoforms: p110α (PI3Kα), p110β (PI3Kβ), p110γ (PI3Kγ), and p110δ (PI3Kδ).[16] In cell-free assays, its half-maximal inhibitory concentration (
IC50) values are exceptionally low, measured at 0.4 nM for PI3Kα, 6.0 nM for PI3Kβ, 5.4 nM for PI3Kγ, and 6.0 nM for PI3Kδ.[3] This inhibitory profile extends to the most common oncogenic mutations of PI3Kα, such as H1047R and E545K, against which it maintains an
IC50 of 0.6 nM.[7]
Concurrently, Gedatolisib is a highly potent inhibitor of the mTOR kinase, with a reported IC50 of 1.6 nM.[3] A key mechanistic feature that distinguishes Gedatolisib from many other mTOR-targeting agents is its equal efficacy in inhibiting both of the distinct multi-protein complexes in which mTOR functions: mTOR Complex 1 (mTORC1) and mTOR Complex 2 (mTORC2).[15]
This dual, comprehensive blockade of both PI3K and mTOR represents a deliberate and rational therapeutic strategy designed to overcome the known limitations of more selective inhibitors. There are three core principles underlying this approach:
The mechanistic rationale for Gedatolisib is strongly supported by a body of preclinical evidence demonstrating its potent biological activity. In vitro, Gedatolisib effectively inhibits the proliferation of various cancer cell lines. For instance, in the MDA-361 (breast cancer) and PC3-MM2 (prostate cancer) cell lines, it demonstrated IC50 values for growth inhibition of 4.0 nM and 13.1 nM, respectively.[7] This anti-proliferative effect is accompanied by clear evidence of on-target pathway modulation; treatment with Gedatolisib leads to a marked suppression of the phosphorylation of key downstream effectors, including AKT, GSK3 kinase, and PRAS40, confirming its ability to disrupt the PAM signaling cascade within the cellular context.[7]
Comparative preclinical studies have consistently highlighted the superiority of Gedatolisib's comprehensive blockade strategy. In head-to-head assessments against single-node PAM inhibitors—such as the PI3Kα-specific alpelisib, the pan-AKT inhibitor capivasertib, and the mTORC1-specific everolimus—Gedatolisib exerted significantly greater growth-inhibitory effects in models of gynecologic and breast cancer.[15] This superior activity extended to the inhibition of critical metabolic functions controlled by the PAM pathway, including cellular glucose consumption and lactate production, suggesting a more profound disruption of the metabolic reprogramming that fuels cancer cell growth.[21]
In vivo, Gedatolisib has demonstrated potent anti-tumor efficacy across a variety of xenograft models, including those derived from H1975, BT474, HCT116, and U87MG cell lines.[7] In a particularly compelling experiment using a staged MDA-361 xenograft model, intravenous administration of Gedatolisib at 20 mg/kg on an intermittent schedule (days 1, 5, and 9) was sufficient to cause the regression of large, established tumors (approx. 900 mm³).[7]
However, it is important to note the context-dependent nature of preclinical findings. One study investigating Gedatolisib in a preclinical model of triple-negative breast cancer (TNBC) metastasis reported that the drug, either alone or in combination with chemotherapy, failed to reliably reduce the burden of disseminated tumor cells (DTCs) or prevent the formation of metastases.[3] This result does not contradict the positive clinical data in HR+ breast cancer but rather underscores a critical principle of drug development: the predictive value of a preclinical model is highly dependent on how well it recapitulates the specific biological dependencies of the targeted human disease. The distinct biology of TNBC compared to HR+ breast cancer likely accounts for this discrepancy, reinforcing that while preclinical data provides essential mechanistic support, definitive evidence of therapeutic value must be established in well-designed clinical trials targeting appropriate patient populations.
The clinical evaluation of Gedatolisib has been extensive and strategic, evolving from broad, early-phase exploration across multiple tumor types to a focused, late-stage program designed to establish its role as a cornerstone therapy in major oncological indications. The program reflects a deep understanding of the PAM pathway's role in cancer and a commitment to addressing areas of significant unmet medical need.
Reflecting the ubiquitous role of PAM pathway dysregulation in malignancy, Gedatolisib has been studied in a wide array of solid tumors.[1] The maximum phase of clinical development reached is Phase 3, signifying its progression to pivotal, registration-enabling studies.[1] Early- and mid-stage clinical trials have enrolled patients with a diverse range of cancers, including Breast Cancer, Endometrial Cancer, Ovarian Cancer, Non-Small Cell Lung Cancer (NSCLC), Small Cell Lung Cancer (SCLC), Squamous Cell Carcinoma of the Head and Neck (SCCHN), and Prostate Cancer.[1] While development was discontinued for some indications such as acute myeloid leukemia and colorectal cancer, the program has strategically prioritized diseases with the strongest biological rationale and largest unmet need, most notably breast and prostate cancer.[12]
The centerpiece of Gedatolisib's clinical development is the VIKTORIA program, a pair of large-scale, multinational Phase 3 trials meticulously designed to define its efficacy and safety in HR+/HER2- advanced breast cancer (ABC), the most common subtype of the disease.
The dual VIKTORIA trials represent a highly sophisticated and commercially astute development strategy. They are not merely confirmatory but are designed to capture two distinct and large market segments within HR+/HER2- ABC. VIKTORIA-1 targets the rapidly growing population of patients who have failed first-line CDK4/6 inhibitors, an area where subsequent therapies offer limited benefit. VIKTORIA-2 aims to establish a new, more effective standard of care for high-risk patients in the first-line setting. Success across this program would position Gedatolisib as a foundational PAM pathway inhibitor throughout the HR+/HER2- ABC treatment continuum.
Beyond breast cancer, Celcuity is actively investigating Gedatolisib in other solid tumors where PAM pathway inhibition is a rational therapeutic strategy.
| Trial Identifier | Trial Name/Acronym | Phase | Condition(s) | Intervention(s) | Status |
|---|---|---|---|---|---|
| NCT05501886 | VIKTORIA-1 | 3 | HR+/HER2- Advanced Breast Cancer (post-CDK4/6i) | Gedatolisib + Fulvestrant +/- Palbociclib | Active 24 |
| N/A | VIKTORIA-2 | 3 | HR+/HER2- Advanced Breast Cancer (1st Line, Endocrine Resistant) | Gedatolisib + CDK4/6i + Fulvestrant | Active 24 |
| NCT06190899 | CELC-G-201 | 1/2 | Metastatic Castration-Resistant Prostate Cancer (mCRPC) | Gedatolisib + Darolutamide | Active 24 |
| NCT03698383 | N/A | 2 | HER2+ Metastatic Breast Cancer (PIK3CA-mutant) | Gedatolisib + Trastuzumab | Unknown Status 32 |
| NCT03065062 | N/A | 1 | Advanced Solid Tumors (Sq. Lung, Pancreatic, H&N) | Gedatolisib + Palbociclib | Temporarily Closed to Accrual 24 |
| NCT03243331 | N/A | 1 | Metastatic Triple-Negative Breast Cancer | Gedatolisib + PTK7-ADC | Completed 34 |
| NCT02626507 | N/A | 1b | ER+/HER2- Breast Cancer (Neoadjuvant) | Gedatolisib + Palbociclib + Fulvestrant | Completed 35 |
| NCT01920061 | N/A | 1 | Advanced Solid Tumors | Gedatolisib + Docetaxel / Cisplatin / Dacomitinib | Completed 36 |
The clinical value of Gedatolisib is defined by the robust efficacy and favorable safety data generated from its comprehensive trial program. The results from the pivotal VIKTORIA-1 study, in particular, represent a landmark achievement in the treatment of advanced breast cancer.
Celcuity has reported topline results from a pre-planned interim analysis of the PIK3CA wild-type cohort of the VIKTORIA-1 trial, and the findings are exceptionally strong and clinically transformative.[19] Both Gedatolisib-containing regimens met the primary endpoint, demonstrating statistically significant and clinically meaningful improvements in progression-free survival (PFS), as determined by Blinded Independent Central Review (BICR), compared to fulvestrant monotherapy.[37]
These results are not merely incremental; they are paradigm-shifting for a patient population with a dire prognosis. Patients with HR+/HER2- ABC who progress on first-line CDK4/6 inhibitor therapy, particularly those with PIK3CA wild-type tumors, have very few effective treatment options. Subsequent endocrine-based therapies typically offer a median PFS of only 2 to 4 months.[18] The VIKTORIA-1 data, showing an incremental PFS benefit of 7.3 months for the triplet and 5.4 months for the doublet over standard therapy, represent a monumental advance. The hazard ratio of 0.24 achieved by the triplet regimen has been described as the lowest ever reported in a Phase 3 trial for this patient population, positioning Gedatolisib to establish an entirely new standard of care upon its potential approval.[18]
| Treatment Arm | Median PFS (months) | 95% CI for mPFS | Hazard Ratio (HR) vs. Fulvestrant | 95% CI for HR | p-value |
|---|---|---|---|---|---|
| Gedatolisib Triplet | 9.3 | 7.2–16.6 | 0.24 | 0.17–0.35 | <0.0001 |
| Gedatolisib Doublet | 7.4 | 5.5–9.9 | 0.33 | 0.24–0.48 | <0.0001 |
| Fulvestrant Monotherapy | 2.0 | 1.8–2.3 | N/A | N/A | N/A |
| Data sourced from 19 |
The clinical utility of a therapeutic agent is determined by the balance of its efficacy and tolerability. In this regard, Gedatolisib has demonstrated a highly favorable and manageable safety profile that sets it apart from other inhibitors of the PAM pathway.[37]
In a Phase 1b study that established the dose for the VIKTORIA-1 trial, the most frequently reported Grade 3-4 treatment-related adverse events (TRAEs) for the triplet combination were hematologic and dermatologic, including neutropenia (63%), stomatitis (inflammation of the mouth, 27%), and rash (20%).[41] These are known on-target effects of PI3K and mTOR inhibition and were considered manageable with standard supportive care.
Critically, the incidence of severe hyperglycemia, a common and often dose-limiting toxicity for this drug class, was remarkably low. Grade 3-4 hyperglycemia was reported in only 6% of participants in the Phase 1b study.[41] This favorable metabolic profile is a significant advantage over other PAM inhibitors, which can be associated with much higher rates of severe hyperglycemia, often requiring dose modifications or discontinuation.
The most compelling evidence of Gedatolisib's excellent tolerability comes from the pivotal VIKTORIA-1 trial, where the treatment discontinuation rate due to adverse events was less than 4% for both the triplet and doublet regimens.[18] This rate is not only lower than what was observed in the earlier Phase 1b study but is also reported to be lower than that of any currently approved combination therapy in a Phase 3 trial for HR+/HER2- ABC.[37] This exceptionally low discontinuation rate is a powerful testament to the drug's therapeutic index. It indicates that patients are able to remain on this highly effective therapy for extended periods, allowing them to derive the maximum possible clinical benefit. This superior safety profile is a major competitive advantage and a key driver of Gedatolisib's potential for widespread clinical adoption.
While the primary focus is on breast cancer, promising signals of activity have emerged from studies in other malignancies.
The transition of Gedatolisib from a promising clinical candidate to a potential standard-of-care therapy is being shaped by a strategic regulatory approach, a strong intellectual property foundation, and a clear corporate vision.
The U.S. Food and Drug Administration (FDA) has recognized the significant potential of Gedatolisib through the granting of multiple special designations, which are intended to expedite the development and review of drugs that address serious conditions and fill an unmet medical need.
This trio of designations represents the FDA's strongest acknowledgment of a drug's potential clinical importance. The acceptance into the RTOR program, in particular, signals that the agency views the VIKTORIA-1 data as highly compelling and recognizes the urgent need for better therapies in this patient population. Celcuity plans to complete its rolling NDA submission to the FDA in the fourth quarter of 2025, suggesting a potential approval on an accelerated timeline.[12]
While the provided documentation does not indicate that a Marketing Authorisation Application (MAA) has been submitted to the European Medicines Agency (EMA) for Gedatolisib, the regulatory environment in the European Union appears favorable for novel PAM pathway inhibitors.[16] The EMA has recently authorized other targeted agents for HR+ breast cancer that modulate this pathway, including the AKT inhibitor capivasertib (Truqap) and the PI3Kα inhibitor inavolisib (Itovebi).[46]
These recent approvals establish a clear regulatory precedent and demonstrate the EMA's acceptance of the PAM pathway as a valid and important therapeutic target in breast cancer. Celcuity's corporate communications have identified the EMA as a target regulatory body for future submissions.[50] Given the strength and magnitude of the VIKTORIA-1 data, a future MAA submission for Gedatolisib would be expected to receive serious consideration, suggesting a high probability of success in securing marketing authorization in the EU.
Gedatolisib is the lead therapeutic candidate for Celcuity Inc., a clinical-stage biotechnology company based in Minneapolis that is focused on the development of targeted cancer therapies.[11] The compound, originally known as PF-05212384, was discovered and initially developed by Wyeth and later Pfizer.[12] In April 2021, Celcuity acquired the exclusive global development and commercialization rights to Gedatolisib from Pfizer.[53] Celcuity's interest in the asset was driven by insights from its proprietary CELsignia platform technology, which identified Gedatolisib as having a superior ability to inhibit PI3K-driven signaling compared to other agents evaluated.[53]
Celcuity has established a robust intellectual property portfolio to protect its lead asset. The company holds multiple patents covering Gedatolisib's composition of matter and various formulations. Most significantly, in July 2025, the U.S. Patent and Trademark Office issued a new patent (U.S. Patent No. 12,350,276) specifically covering the clinical dosing regimen of Gedatolisib in ER+/HER2- breast cancer patients. This key patent extends Celcuity's market exclusivity for Gedatolisib in the United States into 2042, providing a long and secure commercial runway post-approval, protected from generic competition.[11]
Gedatolisib's value proposition is best understood by contextualizing its unique attributes against the landscape of existing and other investigational therapies targeting the PAM pathway. Its differentiation is evident in its mechanism, clinical efficacy, and safety profile.
The first generation of approved PAM pathway inhibitors for breast cancer includes the PI3Kα-specific inhibitor alpelisib and the mTORC1 inhibitor everolimus. While these agents established the clinical validity of targeting the pathway, their benefits have been modest and often accompanied by significant toxicities and the rapid development of resistance.
Numerous preclinical studies have directly compared Gedatolisib to these single-node inhibitors, as well as the investigational AKT inhibitor capivasertib.[15] The results of these studies are consistent and compelling:
Gedatolisib represents a mechanistically advanced, second-generation approach to PAM pathway inhibition. Its ability to comprehensively block multiple nodes simultaneously appears to successfully overcome the primary limitations of its predecessors. The landmark success of Gedatolisib in the PIK3CA wild-type cohort of the VIKTORIA-1 trial provides the ultimate clinical validation of this mechanistic superiority, demonstrating profound efficacy in a patient population where a PI3Kα-specific inhibitor would not be indicated.
The therapeutic concept of dual PI3K/mTOR inhibition has been pursued for over a decade, with several molecules entering clinical development. This class includes compounds such as BEZ235 (dactolisib), samotolisib, and bimiralisib.[55] However, translating this concept into a clinically successful drug has proven to be exceptionally challenging. Many of these earlier candidates were hampered by an unfavorable therapeutic index, exhibiting either insufficient efficacy at tolerable doses or unacceptable toxicity at effective doses. Consequently, many have been discontinued for certain indications or have failed to advance to late-stage development.[12]
Gedatolisib is the first and only dual PI3K/mTOR inhibitor to report unequivocally positive, practice-changing results from a large, randomized Phase 3 trial.[19] Furthermore, its NDA is the first for any drug in this class to be accepted for review by the FDA.[26] Gedatolisib's success in the VIKTORIA-1 trial, which demonstrated both profound efficacy and excellent tolerability, marks a significant breakthrough for the entire class. It is therefore positioned not merely as another agent in the category, but as the undisputed class leader, poised to be the first to achieve regulatory approval and establish the clinical utility of comprehensive PAM pathway blockade.
| Drug Name (Generic) | Target(s) | Highest Development Phase / Approval Status | Key Indication (Breast Cancer) | Key Differentiating Feature | Noteworthy Class-Related Toxicities |
|---|---|---|---|---|---|
| Gedatolisib | Pan-PI3K + mTORC1/2 | Phase 3 / NDA Submitted | HR+/HER2- ABC, post-CDK4/6i | Efficacy in PIK3CA-WT; Favorable safety profile; IV admin | Stomatitis, Rash, Neutropenia, low rate of severe Hyperglycemia |
| Alpelisib | PI3Kα | Approved | HR+/HER2- ABC, PIK3CA-mutant | PIK3CA mutation-specific | Hyperglycemia, Rash, Diarrhea |
| Everolimus | mTORC1 | Approved | HR+/HER2- ABC | Established but modest benefit | Stomatitis, Pneumonitis, Hyperglycemia |
| Capivasertib | Pan-AKT | Approved (EU/other) | HR+/HER2- ABC, PIK3CA/AKT1/PTEN-altered | Targets AKT node | Diarrhea, Rash, Hyperglycemia |
| Dactolisib (BEZ235) | Pan-PI3K + mTOR | Phase 1/2 (Discontinued for some tumors) | N/A | Early development, oral | N/A |
Gedatolisib has emerged as a highly potent, first-in-class, dual pan-PI3K and mTORC1/2 inhibitor that has unequivocally demonstrated its potential to transform the treatment of advanced solid tumors. The results from the pivotal Phase 3 VIKTORIA-1 trial are nothing short of practice-changing, showing an unprecedented improvement in progression-free survival for patients with HR+/HER2-, PIK3CA wild-type advanced breast cancer who have progressed on prior CDK4/6 inhibitor therapy. This landmark achievement addresses a large and growing patient population with a significant unmet medical need.
The drug's unique mechanism of comprehensive PAM pathway blockade appears to be the key to its success, allowing it to overcome the adaptive resistance mechanisms and dose-limiting toxicities that have challenged previous generations of more selective PAM pathway inhibitors. This is further substantiated by its highly favorable safety profile, characterized by a remarkably low rate of treatment discontinuation due to adverse events. With an accelerated regulatory pathway established with the U.S. FDA and a strong intellectual property position extending into the 2040s, Gedatolisib is firmly positioned to become a new standard of care and a cornerstone therapy in oncology.
While the current data are compelling, the clinical story of Gedatolisib is still unfolding, with several key developments on the horizon.
Looking further ahead, long-term questions will focus on the optimal sequencing of Gedatolisib with other therapies, the identification of novel biomarkers beyond PIK3CA status that may predict response or resistance, and the exploration of new rational combination strategies to further enhance its anti-tumor activity. The successful journey of Gedatolisib, from a licensed asset to a potential blockbuster therapeutic, exemplifies a well-executed clinical development strategy rooted in strong scientific rationale and targeted precisely at an area of profound clinical need.
Published at: September 25, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.