Small Molecule
C30H23N5O
915019-65-7
Executive Summary
Dactolisib, also known by its developmental code BEZ235, is a synthetic, orally bioavailable imidazoquinoline derivative that represents a pioneering effort in the therapeutic targeting of the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling pathway. As a first-in-class, dual ATP-competitive inhibitor, Dactolisib was designed to simultaneously block all four Class I PI3K isoforms (α, β, γ, δ) and both mTOR complexes (mTORC1 and mTORC2). This mechanism was predicated on the robust scientific rationale that comprehensive pathway blockade would overcome the intrinsic feedback loops that limit the efficacy of single-agent mTOR inhibitors, thereby offering a more potent and durable antineoplastic effect.
Preclinical investigations revealed Dactolisib to be an exceptionally potent compound. In cell-free assays and across a diverse panel of cancer cell lines, it demonstrated low-nanomolar inhibitory activity, leading to profound downstream effects including cell cycle arrest, induction of apoptosis, and suppression of cellular proliferation and invasion. This potent activity was further validated in numerous in vivo animal models, where oral administration of Dactolisib resulted in significant tumor growth inhibition and, in some cases, regression. Furthermore, the discovery of its potent "off-target" inhibition of key DNA damage response (DDR) kinases, such as ATM, ATR, and DNA-PKcs, provided a strong mechanistic basis for its observed synergy with radiotherapy and chemotherapy in preclinical settings.
Despite this compelling preclinical profile, the clinical development of Dactolisib was marked by a consistent and profound disconnect between its laboratory promise and its performance in human trials. As the first PI3K inhibitor to enter the clinic in 2006, it was evaluated extensively across a wide range of solid tumors and hematological malignancies, both as a monotherapy and in various combinations. However, this broad clinical program was uniformly unsuccessful. Trials were consistently terminated prematurely due to a combination of an unacceptable toxicity profile and a lack of meaningful clinical efficacy. The drug was characterized by severe, dose-limiting adverse events—primarily gastrointestinal and metabolic in nature—and a challenging pharmacokinetic profile marked by low oral bioavailability and high inter-patient variability. This established a narrow, and ultimately non-viable, therapeutic window, where doses required to achieve therapeutic systemic exposure were intolerable to patients.
The development of Dactolisib for oncology was officially discontinued by its sponsor. Subsequent attempts to repurpose the drug at lower doses for non-oncology indications, such as preventing respiratory infections (as RTB101), also failed in late-stage trials. This report provides a comprehensive monograph on Dactolisib, critically analyzing its chemical properties, multifaceted mechanism of action, extensive preclinical and clinical data, and challenging pharmacokinetic and safety profiles. Ultimately, Dactolisib stands as a critical case study in the complexities of cancer drug development. Its failure provided invaluable lessons regarding the importance of therapeutic index, the limitations of preclinical models in predicting human toxicity, and the challenges of broad-spectrum kinase inhibition. The data generated from its development helped illuminate the on-target toxicities of the PI3K/mTOR pathway and guided the field toward the more selective, tolerable, and ultimately more successful next-generation inhibitors that have followed.
This section establishes the fundamental identity of Dactolisib, providing the comprehensive chemical and pharmaceutical information necessary to define the molecule and its properties.
Throughout its lifecycle, Dactolisib has been referenced by multiple names and codes across scientific literature, clinical trial registries, and chemical databases. A clear understanding of this nomenclature is essential for accurate data consolidation.
The International Nonproprietary Name (INN) assigned to the compound is Dactolisib (INN number 9545), which is also its United States Adopted Name (USAN).[1] During its development by Novartis, it was primarily known by the code name NVP-BEZ235, often shortened to BEZ235.[3] Following its out-licensing for non-oncology indications, it was developed by Restorbio under the code RTB101.[3]
The systematic International Union of Pure and Applied Chemistry (IUPAC) name for Dactolisib is 2-methyl-2-[4-(3-methyl-2-oxo-8-quinolin-3-ylimidazo[4,5-c]quinolin-1-yl)phenyl]propanenitrile.[1]
The compound is cataloged across major global databases with consistent identifiers, ensuring its unambiguous tracking. Key identifiers include:
A deprecated CAS number, 1146702-52-4, is also associated with the compound in some databases.[7]
Dactolisib is a complex heterocyclic molecule with properties that influence its formulation, absorption, and interaction with biological systems.
The molecular formula of Dactolisib is C30H23N5O.[6] This corresponds to a molecular weight of approximately 469.54 g/mol, with slight variations reported across sources (e.g., 469.5 g/mol, 469.548 g/mol).[6]
Standardized structural representations are crucial for computational analysis and identification:
From a drug development perspective, Dactolisib's properties align with general guidelines for oral bioavailability, such as Lipinski's Rule of Five. It has 4 hydrogen bond acceptors, 0 hydrogen bond donors, and 3 rotatable bonds, and it does not violate any of Lipinski's rules.[1] Its calculated partition coefficient (XLogP) is approximately 4.92 to 5.2, indicating high lipophilicity.[1]
Dactolisib is formulated as a crystalline solid for research and clinical use.[10] Its solubility is limited in aqueous solutions but can be achieved in organic solvents such as dimethyl sulfoxide (DMSO) and chloroform.[10] For clinical trials, it was developed as an oral agent.[1]
Dactolisib's chemical structure places it within specific chemical and pharmacological classes that define its core activity.
Chemically, it is classified as an imidazoquinoline derivative.[5] This core structure is substituted with a quinolinyl group and a phenylpropanenitrile group. It is also categorized more broadly as a synthetic organic compound, a nitrile, a member of quinolines, a ring assembly, and a member of ureas.[1]
Pharmacologically, its primary classification is as an Antineoplastic Agent.[7] More specifically, it is a Phosphatidylinositide 3-Kinase Inhibitor and an mTOR Inhibitor, reflecting its dual mechanism of action.[11]
To improve its pharmaceutical properties, particularly solubility and stability, Dactolisib has also been formulated as a tosylate salt (Dactolisib Tosylate, CAS: 1028385-32-1).[15] This salt form consists of the Dactolisib free base and p-toluenesulfonic acid.[16]
Table 1: Chemical and Physical Properties of Dactolisib
Property | Value | Source(s) |
---|---|---|
IUPAC Name | 2-methyl-2-[4-(3-methyl-2-oxo-8-quinolin-3-ylimidazo[4,5-c]quinolin-1-yl)phenyl]propanenitrile | 1 |
CAS Number | 915019-65-7 | 7 |
DrugBank ID | DB11651 | 7 |
Molecular Formula | C30H23N5O | 7 |
Molecular Weight | 469.54 g/mol | 8 |
XLogP | 4.92 - 5.2 | 1 |
Hydrogen Bond Acceptors | 4 | 1 |
Hydrogen Bond Donors | 0 | 1 |
Rotatable Bonds | 3 | 1 |
SMILES | CC(C)(C#N)C1=CC=C(C=C1)N2C3=C4C=C(C=CC4=NC=C3N(C2=O)C)C5=CC6=CC=CC=C6N=C5 | 1 |
InChIKey | JOGKUKXHTYWRGZ-UHFFFAOYSA-N | 7 |
The biological activity of Dactolisib is defined by its potent and broad-spectrum inhibition of multiple key cellular kinases. Its primary mechanism involves the simultaneous targeting of the PI3K and mTOR proteins, but its profile is complicated and enhanced by significant activity against kinases involved in the DNA damage response.
The PI3K/AKT/mTOR signaling pathway is a fundamental intracellular cascade that governs a wide range of cellular processes critical for normal physiology and frequently dysregulated in cancer. This pathway integrates signals from growth factors and nutrients to control cell growth, proliferation, survival, metabolism, and angiogenesis.[7] Its constitutive activation, a common event in many human malignancies, is associated with tumor progression, poor prognosis, and resistance to conventional anticancer therapies like chemotherapy and radiotherapy.[5] This central role makes the PI3K/AKT/mTOR axis a highly validated and attractive target for therapeutic intervention.
Dactolisib was designed as a dual inhibitor, a strategy intended to produce a more comprehensive and durable blockade of the PI3K/AKT/mTOR pathway than targeting either PI3K or mTOR alone. The molecule functions as an ATP-competitive inhibitor, meaning it binds to the ATP-binding cleft within the kinase domain of both PI3K and mTOR, thereby preventing the phosphorylation of their respective substrates.[12]
This dual-inhibition strategy is mechanistically sophisticated, designed specifically to overcome a critical limitation of first-generation mTOR inhibitors. The mTOR kinase exists in two distinct complexes, mTORC1 and mTORC2. Early inhibitors, such as rapamycin and its analogues (rapalogs), are allosteric inhibitors that primarily target mTORC1. While this effectively blocks downstream signaling from mTORC1, it also disrupts a negative feedback loop. mTORC1, via its substrate S6K, normally phosphorylates and inhibits insulin receptor substrate 1 (IRS-1), which dampens upstream signaling. When mTORC1 is inhibited by a rapalog, this feedback is lost, leading to increased IRS-1 activity and a compensatory, feedback-driven activation of PI3K and its downstream effector, AKT.[23] This reactivation of AKT can counteract the antiproliferative effects of mTORC1 inhibition and represents a key mechanism of drug resistance.
Dactolisib's design circumvents this problem. By inhibiting PI3K directly, it prevents the upstream feedback activation of AKT that would otherwise be triggered by mTORC1 inhibition. Simultaneously, by inhibiting mTOR, it blocks the downstream effectors of the pathway. This two-pronged attack is theoretically more effective, as it not only blocks signaling at multiple nodes but also preemptively neutralizes a known resistance mechanism, leading to a more profound and sustained suppression of the entire signaling axis.[23]
Dactolisib exhibits potent inhibitory activity against its primary targets with low-nanomolar efficacy in biochemical, cell-free assays. It is considered a pan-Class I PI3K inhibitor, as it targets all four isoforms of this class (p110α, p110β, p110γ, and p110δ).[1] The half-maximal inhibitory concentrations (
IC50) for these isoforms are reported as:
Its potency against p110β is notably lower than against the other three isoforms, but it remains within a biologically active range. Concurrently, Dactolisib is a potent inhibitor of mTOR, with reported IC50 values for mTORC1 (measured via p70S6K activity) ranging from 6 nM to 20.7 nM.[10] Importantly, as an ATP-competitive inhibitor, Dactolisib is capable of inhibiting both mTORC1 and mTORC2 complexes, unlike the rapalogs.[18] The inhibition of mTORC2 is particularly significant, as this complex is responsible for phosphorylating AKT at serine 473, a key step for its full activation. This adds another layer to Dactolisib's comprehensive pathway blockade.
Beyond its intended targets in the PI3K/mTOR pathway, further investigation revealed that Dactolisib is also a potent inhibitor of several key kinases in the DNA Damage Response (DDR) pathway. These kinases belong to the same PI3K-related kinase (PIKK) superfamily as mTOR and share structural similarities in their ATP-binding domains. Dactolisib effectively inhibits ATM (Ataxia-Telangiectasia Mutated), ATR (ATM and Rad3-related), and the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs).[1] The cellular
IC50 for ATR has been measured at 21 nM, a concentration well within the therapeutic range achieved in preclinical models.[1]
This secondary activity has profound functional consequences. ATM and DNA-PKcs are the primary kinases activated in response to DNA double-strand breaks (DSBs), such as those induced by ionizing radiation.[27] They orchestrate the repair of this damage through the nonhomologous end joining (NHEJ) and homologous recombination (HR) pathways. By inhibiting these kinases, Dactolisib effectively cripples the cell's ability to repair radiation-induced DNA damage. This mechanism provides a clear molecular explanation for the powerful radiosensitizing effects of Dactolisib observed in preclinical studies. This effect is distinct from its antiproliferative activity via PI3K/mTOR inhibition and represents a parallel mechanism of action that contributes to its overall antineoplastic profile, particularly in combination with radiotherapy.[27]
The multi-target kinase inhibition by Dactolisib translates into a cascade of downstream effects on cancer cell biology.
Table 2: In Vitro Potency (IC50) of Dactolisib Against Key Kinase Targets
Kinase Target | Reported IC50 (nM) | Assay Type | Source(s) |
---|---|---|---|
PI3K p110α | 4 | Cell-free | 8 |
PI3K p110β | 75 | Cell-free | 8 |
PI3K p110γ | 5 | Cell-free | 8 |
PI3K p110δ | 7 | Cell-free | 8 |
mTOR (mTORC1) | 6 - 20.7 | Cell-free | 21 |
ATR | 21 | Cellular | 1 |
ATM | Potent Inhibitor | Cellular | 27 |
DNA-PKcs | Potent Inhibitor | Cellular | 27 |
Prior to and during its clinical development, Dactolisib was the subject of extensive preclinical investigation that consistently demonstrated potent and broad-spectrum antineoplastic activity. These studies, conducted in a wide range of cancer models, established a strong scientific rationale for its advancement into human trials.
In cell-based assays, Dactolisib displayed robust activity against a diverse array of human cancer cell lines. It effectively reduced cell viability and inhibited proliferation in a dose-dependent manner in models of glioblastoma (A172, SHG44, T98G), breast cancer (MCF-7, MDA-MB-468), leukemia (HL-60/VCR, K562/ADR), lung cancer, and pancreatic cancer, among others.[8] The reported cellular
IC50 values were frequently in the low- to mid-nanomolar range, indicating high potency.[8]
A significant finding from these studies was that Dactolisib's efficacy was largely independent of the specific mutation status of the PI3K pathway within the cancer cells.[13] This suggested that its comprehensive blockade of the pathway could be effective even in tumors without canonical activating mutations in
PIK3CA or loss of PTEN, broadening its potential clinical applicability.
Beyond inhibiting proliferation, Dactolisib also demonstrated the ability to suppress key processes involved in cancer metastasis. Treatment with the compound significantly reduced the migration and invasion of glioblastoma and other cancer cells in wound healing and Transwell invasion assays.[8]
The promising in vitro results were successfully translated into animal models of cancer. When administered orally to mice bearing human tumor xenografts, Dactolisib demonstrated significant antitumor activity. It effectively inhibited tumor growth in models of glioblastoma (U87MG), HER2-amplified breast cancer (BT474), thyroid cancer (K1, C643), and gastric cancer (N87).[8] In some of these models, particularly the BT474 breast cancer model engineered to overexpress a mutant PIK3CA, Dactolisib treatment led not just to growth inhibition but to tumor regression, or shrinkage of established tumors.[13]
Crucially, these in vivo studies confirmed that the observed antitumor effects were accompanied by on-target pharmacodynamic activity. Analysis of tumor tissue from treated animals revealed a significant reduction in the phosphorylation of downstream pathway effectors, such as AKT and the ribosomal protein S6.[24] This demonstrated that orally administered Dactolisib could achieve sufficient concentrations within the tumor microenvironment to effectively engage its molecular targets and inhibit the PI3K/mTOR pathway in a living system.
Given its dual mechanism of inhibiting both proliferation and DNA damage repair, Dactolisib was evaluated in combination with standard cytotoxic therapies. The results were highly synergistic.
In models of glioblastoma (GBM), Dactolisib strongly potentiated the efficacy of the standard-of-care regimen of temozolomide (TMZ) plus radiotherapy (RT). In GBM cell lines, the triple combination of Dactolisib, TMZ, and RT was significantly more effective at reducing cell viability and suppressing cell migration and invasion than TMZ+RT alone.[28] This synergy was confirmed in a more clinically relevant orthotopic rat xenograft model, providing a robust preclinical rationale for testing this combination in GBM patients.[28] This potentiation is a direct functional consequence of Dactolisib's ability to inhibit the DDR kinases ATM and DNA-PKcs, preventing the repair of DNA damage induced by TMZ and RT.
Similar synergistic effects were observed in other cancer types. In lung cancer cell lines, Dactolisib significantly augmented the antitumor effects of the platinum-based chemotherapy agent cisplatin.[30]
The central role of the mTOR pathway in processes beyond cell proliferation, such as autophagy and inflammation, prompted investigation into Dactolisib's potential in non-cancer indications. These studies represented a strategic exploration of whether the drug's mechanism could be leveraged in other disease contexts, potentially at lower, more tolerable doses.
In a transgenic mouse model of Alzheimer's disease (T41 mice), which expresses a mutant amyloid-β precursor protein, a 14-day course of oral Dactolisib (5 mg/kg) yielded promising results. The treatment was found to reduce the social memory impairment observed in these mice. Mechanistically, Dactolisib treatment decreased the activation of microglia, the primary immune cells of the brain, as indicated by a reduced CD68/Iba-1 ratio in the hippocampus. It also restored the levels of the anti-inflammatory cytokine IL-10.[33] These findings suggested that Dactolisib possessed neuroprotective and anti-neuroinflammatory properties, providing a preclinical basis for exploring its use in neurodegenerative diseases. This line of investigation reflects a logical, mechanism-based pivot. The biological processes implicated in neurodegeneration—such as neuroinflammation and the clearance of aggregated proteins via autophagy—are heavily modulated by mTOR. It was plausible that the beneficial effects on these processes could be achieved at concentrations far below those required for cytotoxic effects in oncology, potentially creating a viable therapeutic window where the dose-limiting toxicities seen in cancer trials could be avoided.
The clinical development program for Dactolisib was extensive and ambitious, reflecting the high expectations based on its potent preclinical profile. However, this journey from the laboratory to the clinic was ultimately characterized by a consistent failure to translate preclinical efficacy into meaningful patient benefit, primarily due to an insurmountable toxicity burden.
Dactolisib holds a notable place in the history of targeted therapy as the first PI3K inhibitor to enter human clinical trials, with studies commencing in 2006.[6] Its development program spanned over a decade and explored a wide range of therapeutic areas.
In oncology, it was investigated in numerous Phase I and Phase II trials for the treatment of advanced solid tumors, including breast cancer, renal cell carcinoma (RCC), pancreatic neuroendocrine tumors (pNET), and glioblastoma, among others.[7] It was also studied in hematological malignancies such as acute lymphoid leukemia (ALL).[6] Trials evaluated Dactolisib both as a single agent and in combination with other targeted therapies (e.g., everolimus, trastuzumab, buparlisib) and chemotherapies.[18]
Following its discontinuation in oncology, a second wave of development was initiated by Restorbio, which repurposed the drug (as RTB101) for non-oncology indications, focusing on the prevention of respiratory tract infections in the elderly and as a potential therapy for COVID-19.[3]
The core of Dactolisib's oncology program focused on advanced solid tumors, but these trials consistently encountered insurmountable challenges.
The clinical investigation of Dactolisib in blood cancers was limited and ultimately unsuccessful.
Given the strong preclinical rationale for synergy, Dactolisib was tested in several combination regimens, but this strategy failed to overcome its fundamental limitations. The most notable combination was with the mTORC1 inhibitor everolimus. A Phase Ib study in patients with various advanced solid cancers evaluated this dual-pathway blockade. The trial concluded that the combination had limited efficacy and was poorly tolerated, with severe normal tissue toxicity being a major issue.[6] Other combination trials with agents like the PI3K inhibitor buparlisib, the MEK inhibitor binimetinib, and the HER2-targeted antibody trastuzumab were also conducted but did not lead to a successful therapeutic regimen.[34]
The repurposing of Dactolisib as RTB101 for infectious diseases represented a logical attempt to find a viable therapeutic window at lower doses. An initial Phase IIa randomized, placebo-controlled trial showed that Dactolisib in combination with everolimus decreased the rate of reported infections in an elderly population, providing a signal of potential efficacy.[6] This led to the initiation of larger, late-stage trials. However, a pivotal Phase III study (NCT04668352) aiming to prevent clinically symptomatic respiratory illness in the elderly failed to meet its primary endpoint. Furthermore, a Phase III trial investigating Dactolisib as post-exposure prophylaxis for COVID-19 (NCT04139915) was withdrawn.[2] These failures marked the end of Dactolisib's clinical development.
The developmental history of Dactolisib serves as a stark and compelling case study of the "valley of death" in drug development—the gap between promising preclinical data and successful clinical validation. The drug's failure was not isolated to a specific tumor type, a particular patient population, or a single trial design. Instead, a consistent pattern of failure emerged across its entire clinical program, pointing to fundamental, intrinsic flaws in the molecule's properties.
The preclinical models, while accurately demonstrating on-target pathway inhibition and cellular effects, failed to predict the complex interplay of pharmacokinetics and off-target toxicities that would render the drug's therapeutic index in humans impossibly narrow. The consistent termination of trials for reasons of "toxicity," "lack of efficacy," and "poor tolerability" indicates that Dactolisib could not be administered to patients at a dose high enough or for a duration long enough to achieve the antitumor effects seen in animal models without causing unacceptable harm. This universal failure across diverse clinical settings strongly suggests that the issue lay not with the therapeutic hypothesis but with the drug molecule itself, making Dactolisib a quintessential example of translational failure in oncology.
Table 3: Summary of Major Clinical Trials of Dactolisib (BEZ235/RTB101)
Trial Identifier | Phase | Indication | Intervention | Key Outcome / Reason for Discontinuation | Source(s) |
---|---|---|---|---|---|
N/A | I | Advanced Renal Cell Carcinoma | Dactolisib Monotherapy | Terminated prematurely due to toxicity and lack of clinical efficacy. | 6 |
N/A | II | Advanced Pancreatic Neuroendocrine Tumors (pNET) | Dactolisib Monotherapy | Terminated early due to insufficient normal tissue tolerance (high toxicity). | 6 |
NCT01658436 | II | Everolimus-Resistant pNET | Dactolisib Monotherapy | Poorly tolerated; did not meet endpoint to proceed to Stage 2. | 39 |
NCT01508104 | Ib | Advanced Solid Malignancies | Dactolisib + Everolimus | Limited efficacy and tolerance; severe normal tissue toxicity. | 6 |
NCT01471847 | Ib/II | HER2+ Advanced Breast Cancer | Dactolisib + Trastuzumab | Completed, but did not support further development. | 40 |
N/A | I | Relapsed/Refractory Acute Leukemia | Dactolisib Monotherapy | Initiated in 2012; no results published, presumed terminated. | 6 |
NCT03373903 | IIa | Respiratory Tract Infections (Elderly) | Dactolisib + Everolimus | Decreased rate of reported infections, providing initial proof-of-concept. | 6 |
NCT04668352 | III | Respiratory Tract Infections (Elderly) | Dactolisib (as RTB101) | Failed to meet its primary endpoint. | 2 |
NCT04139915 | III | COVID-19 Post-Exposure Prophylaxis | Dactolisib (as RTB101) | Withdrawn before completion. | 2 |
The pharmacokinetic (PK) properties of Dactolisib, particularly its absorption and bioavailability, proved to be a critical and ultimately insurmountable challenge during its clinical development, contributing significantly to its narrow therapeutic window.
In preclinical literature and summary documents, Dactolisib is consistently described as an "orally bioavailable" compound, a designation supported by its efficacy when administered orally in animal models.[1] However, detailed information regarding its specific metabolic pathways, the enzymes involved (e.g., cytochrome P450 isoforms), and its routes of excretion is not available in the provided documentation.[5] This lack of publicly available, detailed human ADME data is common for investigational drugs that are discontinued early in development.
The transition from preclinical models to human subjects revealed significant issues with Dactolisib's oral absorption and exposure.
A critical aspect of Dactolisib's failure appears to be a vicious cycle involving its gastrointestinal (GI) toxicity and poor bioavailability. The most common and dose-limiting toxicities of Dactolisib were severe GI events, including nausea, vomiting, diarrhea, and mucositis.[18] It is highly plausible that these adverse events directly impaired the drug's absorption from the gut. Furthermore, the low bioavailability itself may have been a consequence of local toxicity to the gastrointestinal lining. This creates a feedback loop: attempts to increase the oral dose to overcome poor absorption would likely exacerbate the very GI side effects that were limiting its entry into the bloodstream. This dynamic makes it nearly impossible to separate the therapeutic dose from the toxic dose, effectively eliminating any viable therapeutic window.[6]
Dactolisib's pharmacokinetic profile and its effects on metabolic pathways also indicated a significant potential for drug-drug interactions.
The clinical development of Dactolisib was ultimately halted not just for a lack of compelling efficacy, but primarily due to an unacceptable and unmanageable safety and tolerability profile. The adverse events observed in human trials were frequent, severe, and consistently led to dose interruptions, reductions, and discontinuations, preventing the administration of a sustained, therapeutically relevant dose.
Across numerous Phase I and II studies, a consistent pattern of toxicities emerged. The most common treatment-related adverse events were predominantly gastrointestinal and metabolic in nature. In a Phase II study in patients with pNET, the most frequently reported all-grade adverse events for Dactolisib were diarrhea (90.3%), stomatitis (74.2%), and nausea (54.8%).[38] Other commonly observed toxicities included fatigue, vomiting, elevated liver enzymes (transaminitis), and hyperglycemia.[18]
What proved to be the insurmountable barrier was the high incidence of severe (Grade 3 or 4) adverse events. In the aforementioned pNET study, 83.9% of patients treated with Dactolisib experienced a Grade 3/4 AE, compared to 71.0% for everolimus. This led to treatment discontinuation due to adverse events in 38.7% of patients on Dactolisib, double the rate seen with everolimus.[38] Similarly, in a trial for everolimus-resistant pNET, 72.7% of patients receiving the 400 mg twice-daily dose and 40.0% of those at a reduced 300 mg dose experienced Grade 3/4 AEs, including hyperglycemia, diarrhea, nausea, and vomiting.[39] This high frequency of severe events underscores the drug's poor tolerability.
Dose-escalation studies designed to find the maximum tolerated dose (MTD) were hampered by the early onset of dose-limiting toxicities (DLTs). In a Phase I study of Dactolisib (as BGT226) given three times weekly, DLTs included Grade 3 nausea/vomiting and diarrhea, which occurred at the 125 mg dose.[43]
The collective evidence from these trials led to a consistent conclusion across multiple independent reports: Dactolisib was "poorly tolerated" and possessed an "unfavorable safety profile".[6] While a lack of efficacy was often cited as a reason for trial termination, a deeper analysis of the data suggests that the prohibitive toxicity profile was the primary driver of its clinical failure. The drug could not be administered at a dose high enough or for a duration long enough to rigorously test its efficacy hypothesis in human patients. The clinical program was effectively halted because the drug was too toxic to administer safely, making the assessment of its potential benefit a secondary and often unreachable goal. The inability to separate the toxic dose from a potentially therapeutic dose meant the drug had no viable therapeutic window in the clinical setting.
Table 4: Incidence of Common Treatment-Emergent Adverse Events (TEAEs) with Dactolisib in a Phase II pNET Trial
Adverse Event | Incidence (All Grades) | Incidence (Grade 3/4) | Source(s) |
---|---|---|---|
Diarrhea | 90.3% | Not Specified | 38 |
Stomatitis | 74.2% | Not Specified | 38 |
Nausea | 54.8% | Not Specified | 38 |
Hyperglycemia | Not Specified | Grade 3/4 in multiple trials | 39 |
Vomiting | Not Specified | Grade 3/4 in multiple trials | 39 |
Total Patients with Grade 3/4 AEs | N/A | 83.9% | 38 |
Discontinuation due to AEs | 38.7% | N/A | 38 |
Note: Data primarily from the randomized Phase II trial in mTOR inhibitor-naïve pNET patients, which provides a direct comparison and clear tolerability data. Specific Grade 3/4 rates for individual AEs were not detailed in the source but were noted as frequent in other trials.
The development and ultimate discontinuation of Dactolisib provide a wealth of information that extends beyond the specifics of a single molecule. Its story offers critical lessons on the challenges of kinase inhibitor development, the evolution of the PI3K/mTOR inhibitor class, and the complex relationship between preclinical potency and clinical viability.
Placing Dactolisib in the context of other dual PI3K/mTOR inhibitors reveals a clear evolutionary trend in the field. Dactolisib's failure was not an isolated event but was representative of the challenges faced by many first-generation, broad-spectrum inhibitors.
The story of Dactolisib is a powerful illustration of the limitations of traditional preclinical drug development models.
The final status of Dactolisib is clear: its development for all oncology indications was halted by Novartis due to its unfavorable safety profile, poor pharmacokinetics, and limited efficacy.[38] Subsequent efforts to repurpose it for non-oncology indications also failed in late-stage clinical trials.[3]
However, Dactolisib should not be viewed merely as a failed drug but rather as a foundational scientific tool and a critical learning experience for the entire field of oncology drug development. As the first PI3K inhibitor to be tested in humans, the vast amount of data generated from its preclinical and clinical studies was invaluable. It provided the first clinical validation that the PI3K/mTOR pathway was indeed "druggable" in humans. The consistent and severe toxicities observed—such as hyperglycemia, rash, and diarrhea—helped to define the "on-target" adverse event profile for this class of inhibitors, informing the safety monitoring plans for all subsequent agents.
The profound failure of this potent, broad-spectrum inhibitor directly highlighted the critical need for greater selectivity. It spurred the rational design and development of the next generation of PI3K pathway inhibitors, which have focused on isoform specificity and improved pharmaceutical properties. In this sense, the failure of Dactolisib was a necessary and informative step on the path toward the successful, approved PI3K inhibitors that are now benefiting patients. It stands as a classic example of a "foundational failure"—a discontinued program whose lessons were instrumental in advancing its field.
Table 5: Comparative Profile of Selected Dual PI3K/mTOR Inhibitors
Inhibitor | Target Profile | Key Indication(s) Studied | Highest Development Phase | Key Clinical Outcome / Status | Distinguishing Features |
---|---|---|---|---|---|
Dactolisib (BEZ235) | Pan-PI3K/mTOR | Advanced Solid Tumors (Breast, pNET, RCC), Leukemia, Infections | Phase III (for infections) | Discontinued. Failed due to high toxicity, poor PK, and limited efficacy. | Oral; First-in-class; High rates of severe GI and metabolic toxicity. |
Gedatolisib (PKI-587) | Pan-PI3K/mTOR | HR+/HER2- Breast Cancer, Prostate Cancer | Phase III | Active. Positive Phase 3 data in breast cancer. | Intravenous; Improved safety profile vs. first-gen agents. |
Bimiralisib (PQR309) | Pan-PI3K/mTOR | Solid Tumors, Lymphoma, Actinic Keratosis | Phase II | Active. Development focusing on intermittent dosing and topical formulation. | Oral/Topical; Intermittent dosing strategy to manage toxicity. |
Voxtalisib (XL765) | Pan-PI3K/mTOR | Lymphoma, High-Grade Glioma | Phase II | Discontinued. Limited efficacy in aggressive lymphomas; development halted. | Oral; Showed some activity in follicular lymphoma but overall profile was unfavorable. |
Published at: September 22, 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.