MedPath

Apitolisib Advanced Drug Monograph

Published:Oct 27, 2025

Generic Name

Apitolisib

Drug Type

Small Molecule

Chemical Formula

C23H30N8O3S

CAS Number

1032754-93-0

Apitolisib (GDC-0980): A Comprehensive Monograph on a Dual PI3K/mTOR Inhibitor in Oncology

Executive Summary

Apitolisib, also known by its developmental codes GDC-0980 and RG7422, is an orally bioavailable, investigational small molecule drug developed by Genentech.[1] It is a potent, dual catalytic inhibitor of Class I phosphatidylinositol 3-kinase (PI3K) and the mammalian target of rapamycin (mTOR) kinase, two central nodes in a critical intracellular signaling pathway.[3] The therapeutic rationale for Apitolisib was to target the PI3K/AKT/mTOR pathway, a network frequently dysregulated in human cancers that drives proliferation, survival, and therapeutic resistance.[5] The strategy of dual inhibition was designed to achieve a more profound and durable pathway blockade than single-agent mTOR inhibitors by simultaneously preventing compensatory feedback loops that can lead to treatment resistance.[1]

In preclinical studies, Apitolisib demonstrated significant promise. It exhibited potent, low-nanomolar inhibition of all four Class I PI3K isoforms and mTOR in vitro and induced robust antineoplastic effects, including cell cycle arrest and apoptosis, across a broad range of cancer cell lines and in vivo xenograft models.[4] This strong preclinical package supported its advancement into clinical trials for various malignancies, including solid tumors, breast cancer, prostate cancer, and renal cell carcinoma.[3]

Despite this early promise, the clinical development of Apitolisib was ultimately constrained by a narrow therapeutic window. The potent, on-target inhibition of the PI3K/mTOR pathway, while effective against tumors, also led to significant mechanism-based toxicities in patients. At clinically effective doses, adverse events such as hyperglycemia, rash, and pneumonitis were common and often severe, limiting the drug's tolerability and leading to high rates of treatment discontinuation.[6] The pivotal Phase II trial in metastatic renal cell carcinoma (mRCC) provided a definitive outcome, demonstrating that Apitolisib was not only more toxic but also significantly less effective than the approved mTORC1 inhibitor everolimus.[12]

Apitolisib represents a classic case study in the "potency-toxicity paradox" that can challenge targeted cancer therapies. While mechanistically sound and preclinically potent, its clinical utility was undermined by the inability to separate profound, systemic target inhibition from unacceptable host toxicity. The lessons learned from its development program have been instrumental, contributing to a broader strategic shift in the field away from pan-inhibitors and toward the development of more selective, isoform-specific PI3K inhibitors with improved safety profiles and a wider therapeutic index.

Molecular Profile and Chemical Synthesis

Identification and Nomenclature

Apitolisib is a synthetic organic compound that has been assigned multiple identifiers throughout its development and in various scientific and chemical databases. Establishing these identifiers is crucial for accurate cross-referencing of research and clinical data. The compound is most commonly known by its generic name, Apitolisib, and its primary developmental code from Genentech, GDC-0980.[4]

The systematic chemical name, according to the International Union of Pure and Applied Chemistry (IUPAC), is (2S)-1-[[2-(2-aminopyrimidin-5-yl)-7-methyl-4-morpholin-4-ylthieno[3,2-d]pyrimidin-6-yl]methyl]piperazin-1-yl]-2-hydroxypropan-1-one.[3] A minor variation, (2S)-1-[[2-(2-amino-5-pyrimidinyl)-7-methyl-4-(4-morpholinyl)thieno[3,2-d]pyrimidin-6-yl]methyl]-1-piperazinyl]-2-hydroxy-1-propanone, is also reported but describes the same chemical entity.[4] A comprehensive list of its key identifiers is provided in Table 1.

Table 1: Chemical and Molecular Identifiers for Apitolisib

Identifier TypeValueSource(s)
Generic NameApitolisib4
Common SynonymsGDC-0980, RG7422, GNE 3904
IUPAC Name(2S)-1-[[2-(2-aminopyrimidin-5-yl)-7-methyl-4-morpholin-4-ylthieno[3,2-d]pyrimidin-6-yl]methyl]piperazin-1-yl]-2-hydroxypropan-1-one3
CAS Number1032754-93-03
DrugBank IDDB121803
PubChem CID2525407116
ChEMBL IDCHEMBL19220943
UNII1C854K1MIJ3
InChIKeyYOVVNQKCSKSHKT-HNNXBMFYSA-N3
SMILESCC1=C(SC2=C1N=C(N=C2N3CCOCC3)C4=CN=C(N=C4)N)CN5CCN(CC5)C(=O)[C@H](C)O3

Physicochemical Properties

Apitolisib is a small molecule with the molecular formula $C_{23}H_{30}N_{8}O_{3}S$ and a molecular weight of 498.6 g/mol.[3] Its structural complexity is captured by standard chemical informatics identifiers, including its SMILES and InChI strings, which provide a linear text-based representation of its three-dimensional structure and stereochemistry.[3]

The compound's solubility characteristics are typical of many orally administered kinase inhibitors, reflecting a balance between the polarity needed for formulation and the lipophilicity required for membrane permeability. It is readily soluble in organic solvents such as dimethylformamide (DMF) and dimethyl sulfoxide (DMSO), with a solubility of 25 mg/ml in both.[4] However, its solubility in aqueous media is poor; for example, in a mixture of DMSO and phosphate-buffered saline (PBS) at pH 7.2 (1:5 ratio), its solubility drops to just 0.15 mg/ml.[4] This property necessitates careful formulation for both preclinical and clinical use to ensure adequate absorption.

Chemical Class and Structure

Apitolisib is classified as a thienopyrimidine, a heterocyclic compound featuring a thiophene ring fused to a pyrimidine ring.[10] This core scaffold is a well-established "privileged structure" in medicinal chemistry, particularly for the design of kinase inhibitors, as it often serves as an effective hinge-binding motif that anchors the molecule within the ATP-binding pocket of the target enzyme.

The molecular architecture of Apitolisib is a highly engineered composite of functional groups, each contributing to its overall pharmacological profile.

  1. Thienopyrimidine Core: Provides the rigid scaffold for interaction with the kinase hinge region.
  2. Morpholino Group: Attached at the C4 position of the pyrimidine ring, this moiety is commonly used to enhance aqueous solubility, improve metabolic stability, and modulate pharmacokinetic properties.[3]
  3. Aminopyrimidine Moiety: Attached at the C2 position, this group likely forms key hydrogen bonds within the active site, contributing to target affinity and selectivity.
  4. Piperazine Linker: A flexible linker connecting the core scaffold to the solvent-exposed region of the binding pocket. This allows for modifications to fine-tune solubility and other properties without disrupting the core binding interactions.[3]
  5. (S)-2-hydroxypropanamide Side Chain: This chiral moiety, attached to the piperazine ring, likely makes specific polar or hydrogen-bonding interactions that enhance binding potency and contribute to the molecule's overall selectivity profile.[3]

The structure is a clear example of rational drug design, where distinct chemical fragments are combined to optimize potency against the intended targets (PI3K and mTOR) while maintaining the drug-like properties required for oral bioavailability and a favorable pharmacokinetic profile.

Synthesis Pathway

The chemical synthesis of Apitolisib is a multi-step process that builds the complex molecule from simpler starting materials, as detailed in the scientific literature.[1] The process involves the sequential construction of the thienopyrimidine core, followed by the attachment of the various side chains.

The synthesis begins with the formation of the core scaffold. Ethyl 3-amino-4-methyl-2-thiophenecarboxylate (I) is reacted with urea (II) via cyclocondensation at high temperature (190 °C) to produce the fused ring system, 7-methylthieno[3,2-d]pyrimidine-2,4-dione (III). This dione intermediate is then activated for subsequent reactions by chlorination with phosphorus oxychloride ($POCl_{3}$) at reflux, yielding the highly reactive 2,4-dichloro-7-methylthieno[3,2-d]pyrimidine (IV).

The next phase involves the selective addition of the morpholine group. Condensation of the dichloro-intermediate (IV) with morpholine (V) in methanol results in the substitution of one chlorine atom to afford 2-chloro-7-methyl-4-(4-morpholinyl)thieno[3,2-d]pyrimidine (VI). To prepare for the attachment of the piperazine side chain, the C6 position of the thiophene ring is functionalized. This is achieved through metalation with butyllithium (BuLi) followed by formylation with DMF, creating the aldehyde derivative (VII).

From this point, two alternative routes can be used to complete the synthesis.

  • Route A: The aldehyde (VII) undergoes reductive amination with N-Boc-piperazine (VIII), a protected form of piperazine, to attach the side chain, yielding intermediate (IX). The tert-butyloxycarbonyl (Boc) protecting group on the piperazine is then cleaved with hydrochloric acid (HCl) to give the free piperazine (X). This intermediate is subsequently acylated with (S)-lactic acid (XI) using a peptide coupling agent like HATU to form the corresponding amide (XII). The final step is a palladium-catalyzed Suzuki coupling reaction between the chloro-amide (XII) and a boronate ester derivative of 2-aminopyrimidine (XIII) to furnish the final product, Apitolisib.
  • Route B: An alternative sequence involves performing the Suzuki coupling earlier in the synthesis. The Boc-protected chloro-intermediate (IX) is first coupled with the boronate ester (XIII) to form the 2-aminopyrimidine derivative (XIV). The Boc group is then removed with HCl to yield the free piperazine intermediate (XV). Finally, this intermediate is condensed with (S)-lactic acid (XI) to give Apitolisib.

Both routes converge on the same final molecule, with the choice of pathway often depending on factors such as reaction yields, purification efficiency, and the availability of starting materials.

The PI3K/AKT/mTOR Signaling Pathway: A Therapeutic Rationale

Pathway Architecture and Function

The phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway is a fundamental intracellular signaling network that governs a multitude of cellular processes, including growth, proliferation, survival, metabolism, and motility.[17] Its central role in normal physiology makes its dysregulation a common and critical event in the development and progression of cancer.

The pathway is typically initiated by the binding of extracellular ligands, such as growth factors (e.g., EGF, IGF) or cytokines, to their cognate receptor tyrosine kinases (RTKs) on the cell surface.[17] This binding event triggers receptor dimerization and autophosphorylation, creating docking sites for signaling proteins containing SH2 domains. Class I PI3Ks, which are heterodimers composed of a p85 regulatory subunit and a p110 catalytic subunit, are recruited to these activated receptors at the plasma membrane.[19] This recruitment relieves the inhibitory constraint of the p85 subunit on the p110 subunit, activating its lipid kinase function. The activated PI3K then catalyzes the phosphorylation of phosphatidylinositol 4,5-bisphosphate ($PIP_{2}$) at the 3'-hydroxyl position of the inositol ring, generating the crucial second messenger phosphatidylinositol 3,4,5-trisphosphate ($PIP_{3}$).[17]

$PIP_{3}$ acts as a docking site for proteins containing pleckstrin homology (PH) domains, bringing them to the inner leaflet of the plasma membrane. Key among these are the serine/threonine kinases AKT (also known as protein kinase B, PKB) and phosphoinositide-dependent kinase 1 (PDK1).[7] At the membrane, PDK1 partially activates AKT via phosphorylation at threonine 308. Full activation of AKT requires a second phosphorylation event at serine 473, which is carried out by the mTOR Complex 2 (mTORC2).[17]

Once fully activated, AKT becomes a central signaling node, phosphorylating a wide array of downstream substrates to promote cell survival (by inhibiting pro-apoptotic proteins like BAD), stimulate cell growth and proliferation (by inhibiting cell cycle inhibitors like p21 and p27), and regulate metabolism.[7]

A major downstream effector of the PI3K/AKT axis is mTOR itself. mTOR is a large serine/threonine kinase that functions as the catalytic core of two distinct multiprotein complexes, mTORC1 and mTORC2.[9]

  • mTORC1: This complex is a master regulator of cell growth and protein synthesis. It is activated downstream of AKT and integrates signals related to growth factors, nutrients (amino acids), and cellular energy status. Activated mTORC1 phosphorylates key substrates such as S6 kinase (S6K) and eukaryotic initiation factor 4E-binding protein 1 (4E-BP1), unleashing mRNA translation and the synthesis of proteins required for cell growth and division.[17]
  • mTORC2: As mentioned, mTORC2 is a key activator of AKT. It is generally considered less sensitive to acute inhibition by rapamycin and its analogs (rapalogs) and plays a critical role in cell survival and the organization of the actin cytoskeleton.[17]

The pathway is tightly regulated by tumor suppressors, most notably PTEN (phosphatase and tensin homolog), a lipid phosphatase that counteracts PI3K activity by dephosphorylating $PIP_{3}$ back to $PIP_{2}$, thereby terminating the signal.[21]

Pathway Dysregulation in Oncogenesis

The PI3K/AKT/mTOR pathway is the most frequently mutated and hyperactivated signaling network in human cancer, making it a highly attractive target for therapeutic intervention.[6] Its constitutive activation provides cancer cells with a significant growth and survival advantage and contributes to resistance against conventional chemotherapy and radiotherapy.[5]

Several distinct molecular mechanisms can lead to the aberrant activation of this pathway in tumors:

  • Activating Mutations in PIK3CA: The gene PIK3CA, which encodes the p110α catalytic subunit of PI3K, is one of the most commonly mutated oncogenes. Hotspot mutations, typically in the helical or kinase domains, render the enzyme constitutively active, leading to constant $PIP_{3}$ production and downstream signaling. These mutations are prevalent in a variety of cancers, including breast, ovarian, endometrial, and head and neck squamous cell carcinomas.[6]
  • Loss of PTEN Function: The tumor suppressor PTEN is frequently inactivated in cancer through mutation, deletion, or epigenetic silencing. Loss of PTEN function prevents the dephosphorylation of $PIP_{3}$, resulting in its accumulation and sustained, ligand-independent activation of AKT and mTOR.[21]
  • Upstream Receptor Activation: Overexpression or activating mutations of upstream RTKs, such as EGFR and HER2, can provide a constant stimulus to PI3K, driving pathway hyperactivation.[9]
  • Mutations in AKT or Other Pathway Components: While less common, activating mutations in AKT itself or inactivating mutations in negative regulators like the TSC1/TSC2 complex can also lead to pathway dysregulation.[21]

The consequence of this sustained signaling is the promotion of multiple cancer hallmarks, including uncontrolled cell proliferation, evasion of apoptosis, increased angiogenesis, enhanced cell motility and metastasis, and altered cellular metabolism (the Warburg effect).[18]

Rationale for Dual Inhibition

The development of Apitolisib was a direct and logical response to the limitations observed with earlier generations of drugs targeting the PI3K/AKT/mTOR pathway. The first clinically approved agents were rapalogs, such as everolimus and temsirolimus, which act as allosteric inhibitors of mTORC1.[6] While these drugs demonstrated clinical activity in certain cancers like renal cell carcinoma and breast cancer, their overall efficacy as single agents was often modest, and resistance frequently developed.[6]

A key biological mechanism underlying this limited efficacy is a powerful negative feedback loop. Under normal conditions, S6K (a downstream target of mTORC1) phosphorylates and inhibits upstream components of the signaling cascade, including insulin receptor substrate (IRS). When mTORC1 is blocked by a rapalog, this negative feedback is relieved. The consequence is an increase in upstream signaling through PI3K, leading to the mTORC2-mediated phosphorylation and hyperactivation of AKT.[6] This rebound activation of AKT, a potent survival signal, can counteract the antiproliferative effects of mTORC1 inhibition and promote therapeutic resistance.[6]

This understanding provided a clear rationale for developing inhibitors that could achieve a more comprehensive blockade of the pathway. The hypothesis was that by simultaneously inhibiting both the upstream driver (PI3K) and the downstream effector (mTOR), it would be possible to prevent this feedback-driven reactivation of AKT. A dual PI3K/mTOR inhibitor like Apitolisib was designed to target three crucial nodes at once: PI3K (blocking $PIP_{3}$ production), mTORC1 (blocking protein synthesis), and mTORC2 (blocking AKT activation).[6] This "triple blockade" was expected to result in a more profound and durable shutdown of the entire signaling axis, leading to superior antitumor activity and the ability to overcome the resistance mechanisms that plagued first-generation mTORC1 inhibitors.[1] This strategic move from single-node to multi-node inhibition represented a rational evolution in therapeutic design. However, this approach also carried an inherent and significant risk: the PI3K/AKT/mTOR pathway is equally critical for normal physiological processes, particularly glucose metabolism regulated by insulin signaling. The very potency and breadth of inhibition that made Apitolisib mechanistically appealing also predisposed it to significant on-target toxicities in healthy tissues, a challenge that would ultimately define its clinical trajectory.

Preclinical Pharmacology and Efficacy

Mechanism of Action and Target Affinity

Apitolisib is characterized as a potent, orally bioavailable, dual catalytic site inhibitor of Class I PI3K and mTOR kinases.[4] Its activity has been extensively quantified in cell-free biochemical assays, which measure the direct interaction of the drug with its purified enzyme targets. These assays confirm that Apitolisib potently inhibits all four isoforms of Class I PI3K (α, β, δ, and γ) as well as the mTOR kinase, with inhibitory concentrations in the low-nanomolar range. This profile validates its classification as a pan-Class I PI3K and mTOR dual inhibitor. The specific inhibitory potencies are summarized in Table 2.

Table 2: In Vitro Inhibitory Potency of Apitolisib against PI3K Isoforms and mTOR

TargetPotency MetricValue (nM)Source(s)
PI3Kα (p110α)$IC_{50}$54
PI3Kβ (p110β)$IC_{50}$274
PI3Kδ (p110δ)$IC_{50}$74
PI3Kγ (p110γ)$IC_{50}$144
mTOR$K_{i}$174

The compound also demonstrates high selectivity. When tested against a large panel of other protein kinases, it was found to be largely inactive, indicating a well-defined target profile.[4] This includes selectivity against other members of the closely related phosphoinositide 3-kinase-related kinase (PIKK) family, such as DNA-dependent protein kinase (DNA-PK), for which its $IC_{50}$ value is significantly higher at 623 nM.[9] This biochemical profile establishes Apitolisib as a potent and selective tool for inhibiting the PI3K/mTOR axis.

In Vitro Antineoplastic Activity

Consistent with its potent biochemical activity, Apitolisib demonstrates broad antineoplastic effects in cultured cancer cells. By inhibiting the PI3K/mTOR pathway, it effectively suppresses the key cellular processes that drive tumorigenesis, resulting in the induction of tumor cell apoptosis (programmed cell death) and the inhibition of cell proliferation.[3]

This activity has been observed across a wide spectrum of cancer cell lines derived from various tumor types, including prostate, breast, non-small cell lung cancer (NSCLC), and glioblastoma.[8] For example, in the PC3 prostate cancer cell line and the MCF7 breast cancer cell line, Apitolisib significantly inhibits cell proliferation with half-maximal inhibitory concentrations ($IC_{50}$) of 307 nM and 255 nM, respectively.[8] Studies in pancreatic cancer cells have shown that Apitolisib can simultaneously activate both apoptosis and autophagy, two critical cellular stress response pathways.[8]

Its effects have also been studied in glioblastoma (GBM), a notoriously difficult-to-treat brain cancer where the PI3K pathway is commonly deregulated. In A-172 and U-118-MG GBM cell lines, Apitolisib induced time- and dose-dependent cytotoxicity and apoptosis.[22] In one experiment, a 48-hour incubation with 20 µM Apitolisib led to apoptosis in 46.47% of A-172 cells.[22] These studies also uncovered a potentially novel mechanism of action in GBM cells, suggesting that Apitolisib may exert an inhibitory effect on PERK, a kinase involved in the unfolded protein response, which could lead to an intensification of protein translation and a subsequent increase in apoptosis.[22]

In Vivo Efficacy in Xenograft Models

The promising in vitro activity of Apitolisib translated into robust antitumor efficacy in preclinical animal models. In mouse xenograft models, where human tumor cells are implanted into immunodeficient mice, orally administered Apitolisib demonstrated significant, dose-dependent tumor growth inhibition.

As a single agent, a low dose of 1 mg/kg was sufficient to cause a significant delay in tumor growth in both PC-3 prostate and MCF-7 breast cancer xenograft models.[8] At its maximum tolerated dose of 7.5 mg/kg, the drug was able to induce tumor stasis (cessation of growth) or even tumor regression.[8] The broad efficacy of the compound was further highlighted in a large study where a 5 mg/kg dose resulted in greater than 50% tumor growth inhibition (TGI) in 15 out of 20 different xenograft models tested.[9] This wide range of activity in preclinical models suggested that Apitolisib could have clinical utility across multiple cancer types.

Furthermore, Apitolisib showed potential for use in combination therapies. In mice bearing tumor xenografts, its administration enhanced the antitumor activity of the widely used chemotherapeutic agent docetaxel, providing a rationale for exploring such combinations in the clinic.[4]

Pharmacokinetics and ADME Profile

A key feature of Apitolisib is its design as an orally available agent, a critical property for convenient, long-term administration in cancer patients.[3] Preclinical pharmacokinetic (PK) studies in mice revealed favorable properties. Following intravenous administration, the drug exhibited low clearance, and after oral administration, it showed dose-proportional exposure over a wide dose range (5 mg/kg to 50 mg/kg).[8] This predictable relationship between dose and exposure is a highly desirable characteristic for a clinical drug candidate, as it simplifies dosing and reduces inter-patient variability.

To formally characterize its behavior in humans, a dedicated Phase I clinical trial (NCT01487239) was conducted. This study was designed to investigate the absorption, metabolism, and excretion (ADME) of a single oral dose of radiolabeled ($[^{14}C]$) Apitolisib in healthy postmenopausal female subjects.[11] Although the specific results of this human ADME study are not detailed in the available documentation, its execution confirms that a thorough characterization of the drug's human pharmacokinetics was a formal part of its development program.

The collective preclinical data for Apitolisib painted the picture of a highly promising drug candidate. It demonstrated potent and selective inhibition of its intended targets, translated this biochemical activity into broad and effective anticancer effects both in vitro and in vivo, and possessed a favorable pharmacokinetic profile suitable for oral administration. This strong preclinical package provided a compelling justification for its advancement into clinical trials. However, a significant disconnect would later emerge between this robust preclinical efficacy and its ultimate clinical performance. This discrepancy highlights a fundamental challenge in oncology drug development: preclinical models, particularly immunodeficient mice with subcutaneous tumors, are excellent for confirming on-target antitumor activity but are often poor predictors of the on-target toxicity that can occur in a complex human system. The systemic, mechanism-based adverse events like hyperglycemia and rash, which arise from inhibiting a pathway vital to normal physiology, are not adequately captured in these efficacy-focused models. The journey of Apitolisib serves as a stark reminder that preclinical efficacy is a necessary but insufficient condition for clinical success, and that predicting the therapeutic window in humans remains a major hurdle.

Clinical Development and Patient Studies

Overview of Clinical Program

The clinical development program for Apitolisib was extensive, investigating the drug as both a monotherapy and in combination with other agents across a range of cancers. The compound advanced to Phase II clinical trials, the maximum phase it achieved, but it has not received regulatory approval from agencies such as the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA).[3] Clinical investigations focused on indications where the PI3K/mTOR pathway is known to be a key driver of disease, including advanced solid tumors, non-Hodgkin's lymphoma, breast cancer, castration-resistant prostate cancer (CRPC), renal cell carcinoma (RCC), and endometrial carcinoma.[3] A summary of the major clinical trials is provided in Table 3.

Table 3: Summary of Major Clinical Trials for Apitolisib

NCT IdentifierPhaseIndication(s)Study DesignKey Outcome/Status
NCT00854152IAdvanced Solid Tumors, Non-Hodgkin's LymphomaDose-escalation to determine MTD and RP2DCompleted; RP2D of 40 mg established. Showed modest but durable antitumor activity with on-target toxicities (hyperglycemia, rash) 6
NCT01442090IIMetastatic Renal Cell Carcinoma (mRCC)Randomized, open-label vs. everolimusCompleted; Apitolisib was less effective and more toxic than everolimus, failing to meet its primary endpoint 12
NCT01485861Ib/IICastration-Resistant Prostate Cancer (CRPC)Combination with abiraterone acetateCompleted; Evaluated safety and efficacy of the combination therapy 11
NCT01455493IIRecurrent/Persistent Endometrial CarcinomaSingle-arm, open-labelCompleted; Reported positive results, particularly in patients with PI3K pathway mutations 11
NCT01254526IMetastatic Breast CancerCombination with paclitaxel +/- bevacizumabCompleted; Assessed safety and pharmacology of the combination regimen 30

Phase I First-in-Human Trial (NCT00854152)

The first-in-human study of Apitolisib was a critical dose-escalation trial involving 120 patients with advanced solid tumors or non-Hodgkin's lymphoma.[6] The primary objectives were to assess the drug's safety, tolerability, maximum tolerated dose (MTD), and recommended Phase II dose (RP2D). Patients received once-daily oral Apitolisib at doses ranging from 2 to 70 mg.[6]

The safety profile was characterized by on-target toxicities directly related to the inhibition of the PI3K/mTOR pathway. Dose-limiting toxicities (DLTs) included Grade 4 fasting hyperglycemia and Grade 3 maculopapular rash, which established the MTD at 50 mg daily.[6] Based on an optimal risk-benefit assessment, the RP2D was determined to be 40 mg once daily on a continuous 28-day cycle.[6] At this dose, the most common Grade 3 or higher adverse events were hyperglycemia (18%), rash (14%), liver dysfunction (12%), diarrhea (10%), and pneumonitis (8%).[6] The risk of pneumonitis prompted the selection of a lower 30 mg dose for a cohort of patients with pleural mesothelioma, who have limited respiratory reserve.[6]

Pharmacokinetic analysis confirmed that Apitolisib exhibited a dose-proportional PK profile, a favorable characteristic.[6] Pharmacodynamic studies provided robust evidence of target engagement in patients. At doses of 16 mg and above, Apitolisib led to significant suppression of phosphorylated AKT (pAkt) levels in platelet-rich plasma (a surrogate tissue) and a greater than 25% decrease in glucose uptake in tumors as measured by fluorodeoxyglucose positron emission tomography (FDG-PET) scans.[6] This confirmed that the drug was biologically active at clinically achievable concentrations.

Despite the toxicity concerns, the study showed evidence of modest but durable single-agent antitumor activity. A total of 10 RECIST partial responses (PRs) were observed, with confirmed responses in patients with peritoneal mesothelioma, pleural mesothelioma, and PIK3CA-mutant squamous cell carcinoma of the head and neck (SCCHN).[6] The activity appeared to be enriched in tumors with alterations in the target pathway; among 14 evaluable patients with known PIK3CA mutations, there were 3 PRs (one confirmed) and 8 instances of stable disease.[6]

Phase II Randomized Trial in Metastatic Renal Cell Carcinoma (mRCC) (NCT01442090)

The most definitive clinical evaluation of Apitolisib came from a randomized Phase II trial that compared it directly against everolimus, the standard-of-care mTORC1 inhibitor, in patients with mRCC.[12] This study was designed to test the hypothesis that dual PI3K/mTOR inhibition would be superior to mTORC1 inhibition alone. Eighty-five patients whose disease had progressed on prior vascular endothelial growth factor (VEGF)-targeted therapy were randomized to receive either Apitolisib 40 mg daily or everolimus 10 mg daily.[2]

The trial failed to meet its primary endpoint and demonstrated that Apitolisib was both less effective and more toxic than everolimus. The comparative efficacy and safety data are summarized in Table 4.

Table 4: Comparison of Efficacy and Key Grade ≥3 Adverse Events: Apitolisib vs. Everolimus in mRCC (NCT01442090)

Endpoint / Adverse EventApitolisib (40 mg)Everolimus (10 mg)
Median Progression-Free Survival (PFS)3.7 months6.1 months
Median Overall Survival (OS)16.5 months22.8 months
Objective Response Rate (ORR)7.1%11.6%
Treatment Discontinuation due to AEs31%12%
Grade ≥3 Hyperglycemia40%9%
Grade ≥3 Rash24%2%
Data sourced from.2

The median PFS was significantly shorter for patients treated with Apitolisib (3.7 months) compared to everolimus (6.1 months), with a hazard ratio of 2.12 indicating a more than two-fold increased risk of progression or death.[2] While not statistically significant, the median OS also trended strongly in favor of everolimus (22.8 vs. 16.5 months).[2]

The inferior efficacy was directly linked to a substantially worse safety profile. Apitolisib was associated with a much higher incidence of high-grade, on-target toxicities, particularly hyperglycemia (40% vs. 9%) and rash (24% vs. 2%).[2] This poor tolerability led to a nearly three-fold higher rate of treatment discontinuation due to adverse events (31% for Apitolisib vs. 12% for everolimus), preventing patients from receiving the drug long enough to derive potential benefit.[2] This outcome demonstrated that the therapeutic hypothesis—that broader pathway inhibition would lead to better outcomes—was defeated not by a lack of biological activity, but by an excess of on-target toxicity, revealing a critically narrow therapeutic window.

Other Key Clinical Investigations

Apitolisib was evaluated in several other clinical contexts. A Phase Ib/II trial (NCT01485861) investigated its use in combination with abiraterone acetate for patients with CRPC previously treated with docetaxel, exploring its potential to overcome resistance to hormonal therapy.[11] In a Phase II single-arm study (NCT01455493) for recurrent or persistent endometrial carcinoma, Apitolisib showed positive results, suggesting potential activity in this malignancy, particularly in tumors harboring PI3K pathway mutations.[11] A Phase I study in metastatic breast cancer (NCT01254526) explored its combination with the chemotherapy agent paclitaxel, with or without the anti-angiogenic agent bevacizumab.[30] While these studies provided additional data on the drug's activity and safety, none led to a pivotal trial or regulatory submission.

Synthesis, Critical Assessment, and Future Outlook

The Challenge of the Therapeutic Window

The clinical development story of Apitolisib is fundamentally a story about the challenge of the therapeutic window. The central reason for its failure was not a lack of potency or an incorrect mechanism of action, but rather an inability to separate its desired on-target effects in tumor cells from its deleterious on-target effects in normal host tissues. The very biochemical potency that made it a promising preclinical candidate—with low-nanomolar $IC_{50}$ values against PI3K and mTOR—became its clinical downfall.[8]

The PI3K/AKT/mTOR pathway is not exclusive to cancer; it is a master regulator of metabolism and growth in healthy cells. Insulin signaling, which is essential for glucose homeostasis, relies heavily on this pathway. Therefore, potent, systemic inhibition of PI3K and mTOR inevitably disrupts normal metabolic function. This was clearly manifested in the high rates of Grade 3-4 hyperglycemia (40% in the mRCC trial) observed in patients treated with Apitolisib.[6] Similarly, the pathway is crucial for the health and maintenance of skin and mucosal tissues, explaining the high incidence of rash and mucositis. The clinical data from the Phase I and Phase II trials demonstrate that the doses required to achieve antitumor activity were inseparable from those that caused these significant toxicities. This narrow therapeutic index meant that many patients could not tolerate the drug long enough for it to be effective, leading to frequent dose reductions and treatment discontinuations, which ultimately compromised its efficacy.[12] Apitolisib worked "too well" in a biological sense, inhibiting the pathway so profoundly that the systemic consequences became clinically untenable.

Comparative Landscape and Developmental Context

Apitolisib's development must be viewed within the broader evolution of PI3K pathway inhibitors. It was conceived as a second-generation agent designed to overcome the limitations of first-generation mTORC1 inhibitors (rapalogs) like everolimus.[6] The head-to-head trial in mRCC was a direct test of this hypothesis, and its decisive failure provided a clear answer: the dual-inhibition strategy, at least as embodied by Apitolisib, was not superior.[12]

The collective experience with Apitolisib and other early-generation pan-PI3K and dual PI3K/mTOR inhibitors helped catalyze a crucial strategic shift in the field. It became apparent that broad, sledgehammer-like inhibition of the entire pathway carried an unacceptable toxicity burden. This realization spurred the development of more refined, isoform-selective inhibitors designed to target specific nodes of the pathway that are preferentially relied upon by cancer cells, while sparing the isoforms more critical for normal physiology.

The clinical and regulatory success of these more selective agents validates this strategic shift. For example, alpelisib (Piqray), a PI3Kα-selective inhibitor, is approved for patients with PIK3CA-mutated, HR-positive breast cancer.[25] Idelalisib (Zydelig), a PI3Kδ-selective inhibitor, is approved for certain hematologic malignancies where the delta isoform is a key driver. These successes demonstrate that a more nuanced approach, targeting specific vulnerabilities, can yield a much better balance of efficacy and safety.

This evolution is even evident within the pipeline of Apitolisib's developer, Genentech. While Apitolisib is no longer in active development, the company has advanced next-generation PI3K inhibitors.[32] A prominent example is inavolisib, a highly potent and selective PI3Kα inhibitor with a unique mechanism that also promotes the degradation of the mutant protein. Inavolisib has received Breakthrough Therapy Designation from the FDA for breast cancer, highlighting its significant promise.[33] This progression from the "broad and potent" GDC-0980 to the "selective and refined" inavolisib serves as a microcosm of the entire pharmaceutical industry's learning curve. The clinical failure of Apitolisib was not a dead end but a critical data point that directly informed the design of superior, more successful subsequent therapies.

Regulatory Status and Future Directions

Apitolisib is an investigational drug and is not approved for any clinical use by the FDA, EMA, or any other global regulatory agency.[3] Given the negative results from its key clinical trials, particularly the pivotal mRCC study, and the emergence of more effective and better-tolerated agents targeting the same pathway, the future prospects for Apitolisib are exceedingly poor. Its development appears to be terminated. While one could hypothesize about niche applications in highly specific, biomarker-defined populations or with novel dosing strategies to mitigate toxicity, it is far more likely that resources will continue to be directed toward the more promising next-generation inhibitors that have already demonstrated a superior therapeutic index.

Concluding Remarks

Apitolisib stands as an important and illustrative chapter in the history of targeted oncology drug development. It was a product of rational design, based on a strong biological hypothesis that dual inhibition of PI3K and mTOR would be a superior therapeutic strategy. Its potent preclinical profile confirmed the power of this approach in non-clinical models. However, its clinical translation was ultimately unsuccessful due to an insurmountable therapeutic index problem. The key lesson from the Apitolisib program is that when targeting pathways that are central to both normal and cancer cell physiology, maximal potency and breadth of inhibition are not always the optimal goals. Clinical success hinges not just on hitting a target, but on hitting it in a manner that patients can tolerate over a prolonged period. The story of Apitolisib provides a critical and enduring lesson for the field, emphasizing that the quest for effective cancer therapies is an iterative process of refining the delicate balance between efficacy and safety, a principle that has guided the successful development of more selective and ultimately more beneficial therapeutic strategies.

Works cited

  1. Apitolisib. Dual PI3K/mTOR inhibitor, Oncolytic., accessed October 27, 2025, https://access.portico.org/Portico/show?viewFile=pdf&auId=pjbf78x5nc5
  2. Randomized Open-Label Phase II Trial of Apitolisib (GDC-0980), a Novel Inhibitor of the PI3K/Mammalian Target of Rapamycin Pathway, Versus Everolimus in Patients With Metastatic Renal Cell Carcinoma - ASCO Publications, accessed October 27, 2025, https://ascopubs.org/doi/abs/10.1200/JCO.2015.64.8808
  3. Apitolisib | C23H30N8O3S | CID 25254071 - PubChem - NIH, accessed October 27, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Apitolisib
  4. GDC-0980 (Apitolisib, RG7422, CAS Number: 1032754-93-0) | Cayman Chemical, accessed October 27, 2025, https://www.caymanchem.com/product/18303/gdc-0980
  5. Definition of apitolisib - NCI Drug Dictionary - National Cancer Institute, accessed October 27, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/apitolisib
  6. Phase I Study of Apitolisib (GDC-0980), Dual Phosphatidylinositol-3-Kinase and Mammalian Target of Rapamycin Kinase Inhibitor, in Patients with Advanced Solid Tumors - AACR Journals, accessed October 27, 2025, https://aacrjournals.org/clincancerres/article/22/12/2874/122000/Phase-I-Study-of-Apitolisib-GDC-0980-Dual
  7. Targeting PI3K/mTOR Signaling in Cancer - AACR Journals, accessed October 27, 2025, https://aacrjournals.org/cancerres/article/71/24/7351/568584/Targeting-PI3K-mTOR-Signaling-in-CancerTargeting
  8. Apitolisib (GDC-0980) | PI3K inhibitor | Mechanism | Concentration - Selleck Chemicals, accessed October 27, 2025, https://www.selleckchem.com/products/GDC-0980-RG7422.html
  9. Apitolisib is an Orally Active Class I PI3K and mTORC1/2 Inhibitor | MedChemExpress, accessed October 27, 2025, https://www.medchemexpress.com/literature/apitolisib-is-an-orally-active-class-i-pi3k-and-mtorc1-2-inhibitor.html
  10. Apitolisib: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed October 27, 2025, https://go.drugbank.com/drugs/DB12180
  11. Apitolisib - Drug Targets, Indications, Patents ... - Patsnap Synapse, accessed October 27, 2025, https://synapse.patsnap.com/drug/ee58b33a37514666abd3eac402f852f2
  12. Randomized Open-Label Phase II Trial of Apitolisib (GDC-0980), a Novel Inhibitor of the PI3K/Mammalian Target of Rapamycin Pathway, Versus Everolimus in Patients With Metastatic Renal Cell Carcinoma | Journal of Clinical Oncology - ASCO Publications, accessed October 27, 2025, https://ascopubs.org/doi/10.1200/JCO.2015.64.8808
  13. A trial looking at apitolisib with everolimus for kidney cancer that has spread (ROVER), accessed October 27, 2025, https://www.cancerresearchuk.org/about-cancer/find-a-clinical-trial/a-trial-looking-at-gdc-0980-with-everolimus-kidney-cancer-that-has-spread-rover
  14. apitolisib | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed October 27, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=7888
  15. Apitolisib (GDC-0980) | Apoptosis Inducer | MedChemExpress, accessed October 27, 2025, https://www.medchemexpress.com/gdc-0980.html
  16. pubchem.ncbi.nlm.nih.gov, accessed October 27, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Apitolisib#:~:text=Apitolisib%20inhibits%20both%20PI3K%20kinase,cancer%20cells%20overexpressing%20PI3K%2FmTOR.
  17. Mechanisms of Resistance to PI3K Inhibitors in Cancer: Adaptive Responses, Drug Tolerance and Cellular Plasticity - MDPI, accessed October 27, 2025, https://www.mdpi.com/2072-6694/13/7/1538
  18. PI3K/Akt/mTOR Pathway and Its Role in Cancer Therapeutics: Are We Making Headway?, accessed October 27, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2022.819128/full
  19. PI3K/AKT/mTOR Signaling Pathway in Breast Cancer: From Molecular Landscape to Clinical Aspects - MDPI, accessed October 27, 2025, https://www.mdpi.com/1422-0067/22/1/173
  20. Targeting PI3K/Akt/mTOR Signaling in Cancer - Frontiers, accessed October 27, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2014.00064/full
  21. Targeting the PI3K/AKT/mTOR Pathway: Biomarkers of Success and Tribulation - PMC, accessed October 27, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3821994/
  22. (PDF) Novel Dual PI3K/mTOR Inhibitor, Apitolisib (GDC-0980), Inhibits Growth and Induces Apoptosis in Human Glioblastoma Cells - ResearchGate, accessed October 27, 2025, https://www.researchgate.net/publication/356264636_Novel_Dual_PI3KmTOR_Inhibitor_Apitolisib_GDC-0980_Inhibits_Growth_and_Induces_Apoptosis_in_Human_Glioblastoma_Cells
  23. PI3K/AKT/mTOR pathway - Wikipedia, accessed October 27, 2025, https://en.wikipedia.org/wiki/PI3K/AKT/mTOR_pathway
  24. (PDF) PI3K/Akt/mTOR Pathway and Its Role in Cancer Therapeutics: Are We Making Headway? - ResearchGate, accessed October 27, 2025, https://www.researchgate.net/publication/359536941_PI3KAktmTOR_Pathway_and_Its_Role_in_Cancer_Therapeutics_Are_We_Making_Headway
  25. Phosphoinositide 3-kinase inhibitor - Wikipedia, accessed October 27, 2025, https://en.wikipedia.org/wiki/Phosphoinositide_3-kinase_inhibitor
  26. Novel Dual PI3K/mTOR Inhibitor, Apitolisib (GDC-0980), Inhibits Growth and Induces Apoptosis in Human Glioblastoma Cells - PubMed, accessed October 27, 2025, https://pubmed.ncbi.nlm.nih.gov/34768941/
  27. Apitolisib Completed Phase 1 Trials for Non-Hodgkin's Lymphoma, Solid Cancers Treatment, accessed October 27, 2025, https://go.drugbank.com/drugs/DB12180/clinical_trials?conditions=DBCOND0043177&phase=1&purpose=treatment&status=completed
  28. Randomized Open-Label Phase II Trial of Apitolisib (GDC-0980), a ..., accessed October 27, 2025, https://ascopubs.org/doi/full/10.1200/JCO.2015.64.8808
  29. NCT01485861 | Study of Ipatasertib or Apitolisib With Abiraterone Acetate Versus Abiraterone Acetate in Participants With Castration-Resistant Prostate Cancer Previously Treated With Docetaxel Chemotherapy | ClinicalTrials.gov, accessed October 27, 2025, https://www.clinicaltrials.gov/study/NCT01485861
  30. Apitolisib Completed Phase 1 Trials for Breast Cancer Treatment | DrugBank Online, accessed October 27, 2025, https://go.drugbank.com/drugs/DB12180/clinical_trials?conditions=DBCOND0028036&phase=1&purpose=treatment&status=completed
  31. FDA approves alpelisib for PIK3CA-related overgrowth spectrum, accessed October 27, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-alpelisib-pik3ca-related-overgrowth-spectrum
  32. Our Pipeline - Genentech, accessed October 27, 2025, https://www.gene.com/medical-professionals/pipeline
  33. FDA Grants Breakthrough Therapy Designation to Genentech's Inavolisib for Advanced Hormone Receptor-Positive, HER2-Negative Breast Cancer With a PIK3CA Mutation, accessed October 27, 2025, https://www.gene.com/media/press-releases/15025/2024-05-20/fda-grants-breakthrough-therapy-designat

Published at: October 27, 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.