Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Xenobiotica ; 54(2): 64-74, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38197324

ABSTRACT

Glioblastoma multiforme (GBM) is the most common primary brain tumour in adults. Available treatments have not markedly improved patient survival in the last twenty years. However, genomic investigations have showed that the PI3K pathway is frequently altered in this glioma, making it a potential therapeutic target.Paxalisib is a brain penetrant PI3K/mTOR inhibitor (mouse Kp,uu 0.31) specifically developed for the treatment of GBM. We characterised the preclinical pharmacokinetics and efficacy of paxalisib and predicted its pharmacokinetics and efficacious dose in humans.Plasma protein binding of paxalisib was low, with the fraction unbound ranging from 0.25 to 0.43 across species. The hepatic clearance of paxalisib was predicted to be low in mice, rats, dogs and humans, and high in monkeys, from hepatocytes incubations. The plasma clearance was low in mice, moderate in rats and high in dogs and monkeys. Oral bioavailability ranged from 6% in monkeys to 76% in rats.The parameters estimated from the pharmacokinetic/pharmacodynamic modelling of the efficacy in the subcutaneous U87 xenograft model combined with the human pharmacokinetics profile predicted by PBPK modelling suggested that a dose of 56 mg may be efficacious in humans. Paxalisib is currently tested in Phase III clinical trials.


Subject(s)
Phosphatidylinositol 3-Kinases , Protein Kinase Inhibitors , Humans , Rats , Mice , Animals , Dogs , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Phosphoinositide-3 Kinase Inhibitors/metabolism , Brain/metabolism , TOR Serine-Threonine Kinases/metabolism
2.
Cancer Discov ; 12(1): 204-219, 2022 01.
Article in English | MEDLINE | ID: mdl-34544753

ABSTRACT

PIK3CA is one of the most frequently mutated oncogenes; the p110a protein it encodes plays a central role in tumor cell proliferation. Small-molecule inhibitors targeting the PI3K p110a catalytic subunit have entered clinical trials, with early-phase GDC-0077 studies showing antitumor activity and a manageable safety profile in patients with PIK3CA-mutant breast cancer. However, preclinical studies have shown that PI3K pathway inhibition releases negative feedback and activates receptor tyrosine kinase signaling, reengaging the pathway and attenuating drug activity. Here we discover that GDC-0077 and taselisib more potently inhibit mutant PI3K pathway signaling and cell viability through unique HER2-dependent mutant p110a degradation. Both are more effective than other PI3K inhibitors at maintaining prolonged pathway suppression. This study establishes a new strategy for identifying inhibitors that specifically target mutant tumors by selective degradation of the mutant oncoprotein and provide a strong rationale for pursuing PI3Kα degraders in patients with HER2-positive breast cancer. SIGNIFICANCE: The PI3K inhibitors GDC-0077 and taselisib have a unique mechanism of action; both inhibitors lead to degradation of mutant p110a protein. The inhibitors that have the ability to trigger specific degradation of mutant p110a without significant change in wild-type p110a protein may result in improved therapeutic index in PIK3CA-mutant tumors.See related commentary by Vanhaesebroeck et al., p. 20.This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Class I Phosphatidylinositol 3-Kinases , Imidazoles , Oxazepines , Phosphoinositide-3 Kinase Inhibitors , Receptor, ErbB-2 , Female , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor/drug effects , Class I Phosphatidylinositol 3-Kinases/genetics , Imidazoles/pharmacology , Imidazoles/therapeutic use , Oxazepines/pharmacology , Oxazepines/therapeutic use , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Receptor, ErbB-2/genetics
3.
Clin Cancer Res ; 26(13): 3135-3144, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32269051

ABSTRACT

PURPOSE: GDC-0084 is an oral, brain-penetrant small-molecule inhibitor of PI3K and mTOR. Because these two targets alter tumor vascularity and metabolism, respectively, we hypothesized multiparametric MR-PET could be used to quantify the response, estimate pharmacokinetic (PK) parameters, and predict progression-free survival (PFS) in patients with recurrent malignant gliomas. PATIENTS AND METHODS: Multiparametric advanced MR-PET imaging was performed to evaluate physiologic response in a first-in-man, multicenter, phase I, dose-escalation study of GDC-0084 (NCT01547546) in 47 patients with recurrent malignant glioma. RESULTS: Measured maximum concentration (C max) was associated with a decrease in enhancing tumor volume (P = 0.0287) and an increase in fractional anisotropy (FA; P = 0.0418). Posttreatment tumor volume, 18F-FDG uptake, Ktrans, and relative cerebral blood volume (rCBV) were all correlated with C max. A linear combination of change in 18F-FDG PET uptake, apparent diffusion coefficient (ADC), FA, Ktrans, vp, and rCBV was able to estimate both C max (R2 = 0.4113; P < 0.0001) and drug exposure (AUC; R2 = 0.3481; P < 0.0001). Using this composite multiparametric MR-PET imaging response biomarker to predict PK, patients with an estimated C max > 0.1 µmol/L and AUC > 1.25 µmol/L*hour demonstrated significantly longer PFS compared with patients with a lower estimated concentration and exposure (P = 0.0039 and P = 0.0296, respectively). CONCLUSIONS: Results from this study suggest composite biomarkers created from multiparametric MR-PET imaging targeting metabolic and/or physiologic processes specific to the drug mechanism of action may be useful for subsequent evaluation of treatment efficacy for larger phase II-III studies.


Subject(s)
Glioma/diagnosis , Glioma/drug therapy , Magnetic Resonance Imaging , Oxazines/pharmacokinetics , Positron-Emission Tomography , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Adult , Aged , Brain/diagnostic imaging , Brain/drug effects , Brain/pathology , Female , Glioma/mortality , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging/methods , Male , Middle Aged , Molecular Targeted Therapy/methods , Neoplasm Grading , Oxazines/administration & dosage , Oxazines/adverse effects , Positron-Emission Tomography/methods , Prognosis , Progression-Free Survival , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Recurrence , Treatment Outcome
4.
Clin Cancer Res ; 26(8): 1820-1828, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31937616

ABSTRACT

PURPOSE: GDC-0084 is an oral, brain-penetrant small-molecule inhibitor of PI3K and mTOR. A first-in-human, phase I study was conducted in patients with recurrent high-grade glioma. PATIENTS AND METHODS: GDC-0084 was administered orally, once daily, to evaluate safety, pharmacokinetics (PK), and activity. Fluorodeoxyglucose-PET (FDG-PET) was performed to measure metabolic responses. RESULTS: Forty-seven heavily pretreated patients enrolled in eight cohorts (2-65 mg). Dose-limiting toxicities included 1 case of grade 2 bradycardia and grade 3 myocardial ischemia (15 mg), grade 3 stomatitis (45 mg), and 2 cases of grade 3 mucosal inflammation (65 mg); the MTD was 45 mg/day. GDC-0084 demonstrated linear and dose-proportional PK, with a half-life (∼19 hours) supportive of once-daily dosing. At 45 mg/day, steady-state concentrations exceeded preclinical target concentrations producing antitumor activity in xenograft models. FDG-PET in 7 of 27 patients (26%) showed metabolic partial response. At doses ≥45 mg/day, a trend toward decreased median standardized uptake value in normal brain was observed, suggesting central nervous system penetration of drug. In two resection specimens, GDC-0084 was detected at similar levels in tumor and brain tissue, with a brain tissue/tumor-to-plasma ratio of >1 and >0.5 for total and free drug, respectively. Best overall response was stable disease in 19 patients (40%) and progressive disease in 26 patients (55%); 2 patients (4%) were nonevaluable. CONCLUSIONS: GDC-0084 demonstrated classic PI3K/mTOR-inhibitor related toxicities. FDG-PET and concentration data from brain tumor tissue suggest that GDC-0084 crossed the blood-brain barrier.


Subject(s)
Brain/metabolism , Glioma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Oxazines/pharmacokinetics , Oxazines/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adult , Aged , Biomarkers, Tumor/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Disease Progression , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Female , Glioma/metabolism , Glioma/pathology , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Patient Safety , Tissue Distribution
5.
Bioorg Med Chem Lett ; 29(16): 2294-2301, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31307887

ABSTRACT

CDK4 and CDK6 are kinases with similar sequences that regulate cell cycle progression and are validated targets in the treatment of cancer. Glioblastoma is characterized by a high frequency of CDKN2A/CCND2/CDK4/CDK6 pathway dysregulation, making dual inhibition of CDK4 and CDK6 an attractive therapeutic approach for this disease. Abemaciclib, ribociclib, and palbociclib are approved CDK4/6 inhibitors for the treatment of HR+/HER2- breast cancer, but these drugs are not expected to show strong activity in brain tumors due to poor blood brain barrier penetration. Herein, we report the identification of a brain-penetrant CDK4/6 inhibitor derived from a literature molecule with low molecular weight and topological polar surface area (MW = 285 and TPSA = 66 Å2), but lacking the CDK2/1 selectivity profile due to the absence of a basic amine. Removal of a hydrogen bond donor via cyclization of the pyrazole allowed for the introduction of basic and semi-basic amines, while maintaining in many cases efflux ratios reasonable for a CNS program. Ultimately, a basic spiroazetidine (cpKa = 8.8) was identified that afforded acceptable selectivity over anti-target CDK1 while maintaining brain-penetration in vivo (mouse Kp,uu = 0.20-0.59). To probe the potency and selectivity, our lead compound was evaluated in a panel of glioblastoma cell lines. Potency comparable to abemaciclib was observed in Rb-wild type lines U87MG, DBTRG-05MG, A172, and T98G, while Rb-deficient cell lines SF539 and M059J exhibited a lack of sensitivity.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Design , Glioblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , MCF-7 Cells , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
6.
Drug Metab Dispos ; 44(12): 1881-1889, 2016 12.
Article in English | MEDLINE | ID: mdl-27638506

ABSTRACT

Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults. Limited treatment options have only marginally impacted patient survival over the past decades. The phophatidylinositol 3-kinase (PI3K) pathway, frequently altered in GBM, represents a potential target for the treatment of this glioma. 5-(6,6-Dimethyl-4-morpholino-8,9-dihydro-6H-[1,4]oxazino[4,3-e]purin-2-yl)pyrimidin-2-amine (GDC-0084) is a PI3K inhibitor that was specifically optimized to cross the blood-brain barrier. The goals of our studies were to characterize the brain distribution, pharmacodynamic (PD) effect, and efficacy of GDC-0084 in orthotopic xenograft models of GBM. GDC-0084 was tested in vitro to assess its sensitivity to the efflux transporters P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) and in vivo in mice to evaluate its effects on the PI3K pathway in intact brain. Mice bearing U87 or GS2 intracranial tumors were treated with GDC-0084 to assess its brain distribution by matrix-assisted laser desorption ionization (MALDI) imaging and measure its PD effects and efficacy in GBM orthotopic models. Studies in transfected cells indicated that GDC-0084 was not a substrate of P-gp or BCRP. GDC-0084 markedly inhibited the PI3K pathway in mouse brain, causing up to 90% suppression of the pAkt signal. MALDI imaging showed GDC-0084 distributed evenly in brain and intracranial U87 and GS2 tumors. GDC-0084 achieved significant tumor growth inhibition of 70% and 40% against the U87 and GS2 orthotopic models, respectively. GDC-0084 distribution throughout the brain and intracranial tumors led to potent inhibition of the PI3K pathway. Its efficacy in orthotopic models of GBM suggests that it could be effective in the treatment of GBM. GDC-0084 is currently in phase I clinical trials.


Subject(s)
Brain Neoplasms/metabolism , Brain/metabolism , Glioblastoma/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain Neoplasms/drug therapy , Cell Line , Cell Line, Tumor , Dogs , Female , Glioblastoma/drug therapy , Glioma/drug therapy , Glioma/metabolism , Humans , Indazoles/metabolism , Indazoles/pharmacology , Madin Darby Canine Kidney Cells , Mice , Mice, Nude , Protein Kinase Inhibitors/pharmacology
7.
ACS Med Chem Lett ; 7(4): 351-6, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27096040

ABSTRACT

Inhibition of phosphoinositide 3-kinase (PI3K) signaling is an appealing approach to treat brain tumors, especially glioblastoma multiforme (GBM). We previously disclosed our successful approach to prospectively design potent and blood-brain barrier (BBB) penetrating PI3K inhibitors. The previously disclosed molecules were ultimately deemed not suitable for clinical development due to projected poor metabolic stability in humans. We, therefore, extended our studies to identify a BBB penetrating inhibitor of PI3K that was also projected to be metabolically stable in human. These efforts required identification of a distinct scaffold for PI3K inhibitors relative to our previous efforts and ultimately resulted in the identification of GDC-0084 (16). The discovery and preclinical characterization of this molecule are described within.

8.
J Med Chem ; 59(3): 985-1002, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26741947

ABSTRACT

Inhibitors of the class I phosphoinositide 3-kinase (PI3K) isoform PI3Kα have received substantial attention for their potential use in cancer therapy. Despite the particular attraction of targeting PI3Kα, achieving selectivity for the inhibition of this isoform has proved challenging. Herein we report the discovery of inhibitors of PI3Kα that have selectivity over the other class I isoforms and all other kinases tested. In GDC-0032 (3, taselisib), we previously minimized inhibition of PI3Kß relative to the other class I insoforms. Subsequently, we extended our efforts to identify PI3Kα-specific inhibitors using PI3Kα crystal structures to inform the design of benzoxazepin inhibitors with selectivity for PI3Kα through interactions with a nonconserved residue. Several molecules selective for PI3Kα relative to the other class I isoforms, as well as other kinases, were identified. Optimization of properties related to drug metabolism then culminated in the identification of the clinical candidate GDC-0326 (4).


Subject(s)
Antineoplastic Agents/pharmacology , Benzoxepins/pharmacology , Drug Design , Imidazoles/pharmacology , Oxazepines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Benzoxepins/chemistry , Benzoxepins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Class I Phosphatidylinositol 3-Kinases , Dogs , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Macaca fascicularis , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Oxazepines/chemistry , Oxazepines/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
9.
Drug Metab Dispos ; 42(7): 1110-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24754926

ABSTRACT

Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults, and the limited available treatment options have not meaningfully impacted patient survival in the past decades. Such poor outcomes can be at least partly attributed to the inability of most drugs tested to cross the blood-brain barrier and reach all areas of the glioma. The objectives of these studies were to visualize and compare by matrix-assisted laser desorption ionization (MALDI) imaging mass spectrometry the brain and tumor distribution of the phosphatidylinositol 3-kinase (PI3K) inhibitors pictilisib (GDC-0941, 2-(1H-indazol-4-yl)-6-(4-methanesulfonyl-piperazin-1-ylmethyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidine) and GNE-317 [5-(6-(3-methoxyoxetan-3-yl)-7-methyl-4-morpholinothieno[3,2-d]pyrimidin-2-yl)pyrimidin-2-amine] in U87 and GS2 orthotopic models of GBM, models that exhibit differing blood-brain barrier characteristics. Following administration to tumor-bearing mice, pictilisib was readily detected within tumors of the contrast-enhancing U87 model whereas it was not located in tumors of the nonenhancing GS2 model. In both GBM models, pictilisib was not detected in the healthy brain. In contrast, GNE-317 was uniformly distributed throughout the brain in the U87 and GS2 models. MALDI imaging revealed also that the pictilisib signal varied regionally by up to 6-fold within the U87 tumors whereas GNE-317 intratumor levels were more homogeneous. Liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) analyses of the nontumored half of the brain showed pictilisib had brain-to-plasma ratios lower than 0.03 whereas they were greater than 1 for GNE-317, in agreement with their brain penetration properties. These results in orthotopic models representing either the contrast-enhancing or invasive areas of GBM clearly demonstrate the need for whole-brain distribution to potentially achieve long-term efficacy in GBM.


Subject(s)
Brain Neoplasms/metabolism , Enzyme Inhibitors/pharmacokinetics , Glioblastoma/metabolism , Indazoles/pharmacokinetics , Phosphoinositide-3 Kinase Inhibitors , Pyrimidines/pharmacokinetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Sulfonamides/pharmacokinetics , Thiophenes/pharmacokinetics , Animals , Enzyme Inhibitors/pharmacology , Female , Humans , Indazoles/pharmacology , Mice , Mice, Nude , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Thiophenes/pharmacology , Tissue Distribution
10.
J Med Chem ; 56(11): 4597-610, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-23662903

ABSTRACT

Dysfunctional signaling through the phosphoinositide 3-kinase (PI3K)/AKT/mTOR pathway leads to uncontrolled tumor proliferation. In the course of the discovery of novel benzoxepin PI3K inhibitors, we observed a strong dependency of in vivo antitumor activity on the free-drug exposure. By lowering the intrinsic clearance, we derived a set of imidazobenzoxazepin compounds that showed improved unbound drug exposure and effectively suppressed growth of tumors in a mouse xenograft model at low drug dose levels. One of these compounds, GDC-0032 (11l), was progressed to clinical trials and is currently under phase I evaluation as a potential treatment for human malignancies.


Subject(s)
Antineoplastic Agents/chemical synthesis , Imidazoles/chemical synthesis , Oxazepines/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Hepatocytes/metabolism , Humans , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Mice , Mice, Nude , Microsomes, Liver/metabolism , Neoplasm Transplantation , Oxazepines/pharmacokinetics , Oxazepines/pharmacology , Structure-Activity Relationship , Transplantation, Heterologous
11.
Bioorg Med Chem Lett ; 23(9): 2606-13, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23540645

ABSTRACT

A series of suitable five-membered heterocyclic alternatives to thiophenes within a thienobenzoxepin class of PI3-kinase (PI3K) inhibitors was discovered. Specific thiazolobenzoxepin 8-substitution was identified that increased selectivity over PI3Kß. PI3Kß-sparing compound 27 (PI3Kß Ki,app/PI3Kα Ki,app=57) demonstrated dose-dependent knockdown of pAKT, pPRAS40 and pS6RP in vivo as well as differential effects in an in vitro proliferation cell line screen compared to pan PI3K inhibitor GDC-0941. A new structure-based hypothesis for reducing inhibition of the PI3K ß isoform while maintaining activity against α, δ and γ isoforms is presented.


Subject(s)
Benzoxepins/chemistry , Enzyme Inhibitors/chemistry , Phosphoinositide-3 Kinase Inhibitors , Thiazoles/chemistry , Benzoxepins/chemical synthesis , Benzoxepins/pharmacology , Binding Sites , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , MCF-7 Cells , Molecular Docking Simulation , Phosphatidylinositol 3-Kinase/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/metabolism , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 23(3): 897-901, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23265894

ABSTRACT

Substructural class effects surrounding replacement of a 'cis' N-methyl aniline amide within potent and selective thienobenzoxepin PI3-kinase inhibitors are disclosed. While a simple aryl to alkyl switch was not tolerated due to differences in preferred amide conformation, heterocyclic amide isosteres with maintained aryl substitution improved potency and metabolic stability at the cost of physical properties. These gains in potency allowed lipophilic deconstruction of the arene to simple branched alkyl substituents. As such, overall lipophilicity-neutral, MW decreases were realized relative to the aniline amide series. The improved properties for lead compound 21 resulted in high permeability, solubility and bioavailability.


Subject(s)
Benzoxepins/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors , Amides/chemical synthesis , Amides/chemistry , Amides/pharmacology , Benzothiazoles/chemistry , Benzoxepins/chemistry , Benzoxepins/pharmacology , Binding Sites , Crystallography, X-Ray , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Models, Molecular , Thiophenes/chemical synthesis , Thiophenes/chemistry , Thiophenes/pharmacology
13.
J Med Chem ; 55(18): 8007-20, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22946614

ABSTRACT

Inhibition of phosphoinositide 3-kinase (PI3K) signaling through PI3Kα has received significant attention for its potential in cancer therapy. While the PI3K pathway is a well-established and widely pursued target for the treatment of many cancer types due to the high frequency of abnormal PI3K signaling, glioblastoma multiforme (GBM) is particularly relevant because the pathway is implicated in more than 80% of GBM cases. Herein, we report the identification of PI3K inhibitors designed to cross the blood-brain barrier (BBB) to engage their target where GBM tumors reside. We leveraged our historical experience with PI3K inhibitors to identify correlations between physicochemical properties and transporter efflux as well as metabolic stability to focus the selection of molecules for further study.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Design , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dogs , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Hydrogen Bonding , Madin Darby Canine Kidney Cells , Mice , Permeability , Surface Properties
14.
Clin Cancer Res ; 18(22): 6239-48, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22992516

ABSTRACT

PURPOSE: Glioblastoma (GBM), the most common primary brain tumor in adults, presents a high frequency of alteration in the PI3K pathway. Our objectives were to identify a dual PI3K/mTOR inhibitor optimized to cross the blood-brain barrier (BBB) and characterize its brain penetration, pathway modulation in the brain and efficacy in orthotopic xenograft models of GBM. EXPERIMENTAL DESIGN: Physicochemical properties of PI3K inhibitors were optimized using in silico tools, leading to the identification of GNE-317. This compound was tested in cells overexpressing P-glycoprotein (P-gp) or breast cancer resistance protein (BCRP). Following administration to mice, GNE-317 plasma and brain concentrations were determined, and phosphorylated biomarkers (pAkt, p4EBP1, and pS6) were measured to assess PI3K pathway suppression in the brain. GNE-317 efficacy was evaluated in the U87, GS2, and GBM10 orthotopic models of GBM. RESULTS: GNE-317 was identified as having physicochemical properties predictive of low efflux by P-gp and BCRP. Studies in transfected MDCK cells showed that GNE-317 was not a substrate of either transporter. GNE-317 markedly inhibited the PI3K pathway in mouse brain, causing 40% to 90% suppression of the pAkt and pS6 signals up to 6-hour postdose. GNE-317 was efficacious in the U87, GS2, and GBM10 orthotopic models, achieving tumor growth inhibition of 90% and 50%, and survival benefit, respectively. CONCLUSIONS: These results indicated that specific optimization of PI3K inhibitors to cross the BBB led to potent suppression of the PI3K pathway in healthy brain. The efficacy of GNE-317 in 3 intracranial models of GBM suggested that this compound could be effective in the treatment of GBM.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Blood-Brain Barrier/metabolism , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Thiophenes/pharmacokinetics , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Capillary Permeability , Cell Line , Cell Membrane Permeability , Dogs , Female , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Mice , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Thiophenes/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Drug Metab Dispos ; 40(9): 1785-96, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22696419

ABSTRACT

(S)-1-{4-[2-(2-Amino-pyrimidin-5-yl)-7-methyl-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]-piperazin-1-yl}-2-hydroxy-propan-1-one (GDC-0980) is a potent and selective inhibitor of phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin, two key components of the PI3K pathway, the deregulation of which is associated with the development of many cancers. The objectives of these studies were to characterize the absorption and disposition of GDC-0980 and assess its efficacy in an MCF7-neo/HER2 human breast cancer xenograft model in immunocompromised mice. Studies in parental Madin-Darby canine kidney cells indicated that GDC-0980 had high permeability (P(app) = 18 × 10⁻6 cm/s), suggesting good absorption potential. However, it was found to be a P-glycoprotein and breast cancer resistance protein substrate in transfected cells and in knockout mice studies. Plasma protein binding was low, with the fraction unbound ranging from 29 to 52% across species. GDC-0980 hepatic clearance (CL) was predicted to be low in all of the species tested from hepatocyte incubations. The plasma CL of GDC-0980 was low in mouse (6.30 ml · min⁻¹ · kg⁻¹), rat (15.4 ml · min⁻¹ · kg⁻¹), and dog (6.37 ml · min⁻¹ · kg⁻¹) and moderate in cynomolgus monkey (18.9 ml · min⁻¹ · kg⁻¹). Oral bioavailability ranged from 14.4% in monkey to 125% in dog. Predicted human plasma CL and volume of distribution using allometry were 5.1 ml · min⁻¹ · kg⁻¹ and 1.8 l/kg, respectively. Parameters estimated from the pharmacokinetic/pharmacodynamic modeling of the MCF7-neo/HER2 xenograft data indicated that the GDC-0980 plasma concentration required for tumor stasis was approximately 0.5 µM. These parameters, combined with the predicted human pharmacokinetic profile, suggested that 55 mg once daily may be a clinically efficacious dose. GDC-0980 preclinical characterization and the predictions of its human properties supported its clinical development; it is currently in Phase II clinical trials.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/drug therapy , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Intestinal Absorption , Models, Biological , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Area Under Curve , Biological Availability , Biotransformation , Brain/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/blood , Cell Membrane Permeability , Dogs , Drug Dosage Calculations , Female , Half-Life , Hepatocytes/metabolism , Humans , Injections, Intravenous , Liver/metabolism , MCF-7 Cells , Macaca fascicularis , Madin Darby Canine Kidney Cells , Male , Metabolic Clearance Rate , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinase/metabolism , Protein Binding , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/blood , Pyrimidines/administration & dosage , Pyrimidines/blood , Rats , Rats, Sprague-Dawley , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Species Specificity , TOR Serine-Threonine Kinases/metabolism , Tissue Distribution , Transfection , Xenograft Model Antitumor Assays
16.
J Med Chem ; 54(22): 7815-33, 2011 Nov 24.
Article in English | MEDLINE | ID: mdl-21985639

ABSTRACT

Of the four class I phosphoinositide 3-kinase (PI3K) isoforms, PI3Kα has justly received the most attention for its potential in cancer therapy. Herein we report our successful approaches to achieve PI3Kα vs PI3Kß selectivity for two chemical series. In the thienopyrimidine series of inhibitors, we propose that select ligands achieve selectivity derived from a hydrogen bonding interaction with Arg770 of PI3Kα that is not attained with the corresponding Lys777 of PI3Kß. In the benzoxepin series of inhibitors, the selectivity observed can be rationalized by the difference in electrostatic potential between the two isoforms in a given region rather than any specific interaction.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Models, Molecular , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Benzoxepins/chemistry , Benzoxepins/pharmacokinetics , Benzoxepins/pharmacology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/chemistry , Crystallography, X-Ray , Drug Design , Drug Screening Assays, Antitumor , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Bonding , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Mice , Piperazines/chemistry , Piperazines/pharmacokinetics , Piperazines/pharmacology , Protein Conformation , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Structure-Activity Relationship
17.
J Med Chem ; 54(21): 7579-87, 2011 Nov 10.
Article in English | MEDLINE | ID: mdl-21981714

ABSTRACT

The discovery of 2 (GDC-0980), a class I PI3K and mTOR kinase inhibitor for oncology indications, is described. mTOR inhibition was added to the class I PI3K inhibitor 1 (GDC-0941) scaffold primarily through the substitution of the indazole in 1 for a 2-aminopyrimidine. This substitution also increased the microsomal stability and the free fraction of compounds as evidenced through a pairwise comparison of molecules that were otherwise identical. Highlighted in detail are analogues of an advanced compound 4 that were designed to improve solubility, resulting in 2. This compound, is potent across PI3K class I isoforms with IC(50)s of 5, 27, 7, and 14 nM for PI3Kα, ß, δ, and γ, respectively, inhibits mTOR with a K(i) of 17 nM yet is highly selective versus a large panel of kinases including others in the PIKK family. On the basis of the cell potency, low clearance in mouse, and high free fraction, 2 demonstrated significant efficacy in mouse xenografts when dosed as low as 1 mg/kg orally and is currently in phase I clinical trials for cancer.


Subject(s)
Antineoplastic Agents/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Pyrimidines/chemical synthesis , TOR Serine-Threonine Kinases/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Humans , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Mice , Mice, Nude , Microsomes, Liver/metabolism , Models, Molecular , Neoplasm Transplantation , Protein Conformation , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous
18.
Mol Cancer Ther ; 10(12): 2426-36, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21998291

ABSTRACT

Alterations of the phosphoinositide-3 kinase (PI3K)/Akt signaling pathway occur broadly in cancer via multiple mechanisms including mutation of the PIK3CA gene, loss or mutation of phosphatase and tensin homolog (PTEN), and deregulation of mammalian target of rapamycin (mTOR) complexes. The dysregulation of this pathway has been implicated in tumor initiation, cell growth and survival, invasion and angiogenesis, thus, PI3K and mTOR are promising therapeutic targets for cancer. We discovered GDC-0980, a selective, potent, orally bioavailable inhibitor of Class I PI3 kinase and mTOR kinase (TORC1/2) with excellent pharmacokinetic and pharmaceutical properties. GDC-0980 potently inhibits signal transduction downstream of both PI3K and mTOR, as measured by pharmacodynamic (PD) biomarkers, thereby acting upon two key pathway nodes to produce the strongest attainable inhibition of signaling in the pathway. Correspondingly, GDC-0980 was potent across a broad panel of cancer cell lines, with the greatest potency in breast, prostate, and lung cancers and less activity in melanoma and pancreatic cancers, consistent with KRAS and BRAF acting as resistance markers. Treatment of cancer cell lines with GDC-0980 resulted in G1 cell-cycle arrest, and in contrast to mTOR inhibitors, GDC-0980 induced apoptosis in certain cancer cell lines, including those with direct pathway activation via PI3K and PTEN. Low doses of GDC-0980 potently inhibited tumor growth in xenograft models including those with activated PI3K, loss of LKB1 or PTEN, and elicited an exposure-related decrease in PD biomarkers. These preclinical data show that GDC-0980 is a potent and effective dual PI3K/mTOR inhibitor with promise for the clinic.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Neoplasms/drug therapy , Pyrimidines/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , HCT116 Cells , Humans , Mice , Models, Theoretical , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/classification , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/classification , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
19.
Xenobiotica ; 41(12): 1088-99, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21838594

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K) pathway is a major determinant of cell cycling and proliferation. Its deregulation is associated with the development of many cancers. GDC-0941, a potent and selective inhibitor of PI3K, was characterised preclinically in in vitro and in vivo studies. Plasma protein binding was extensive, with free fraction less than 7%, and blood-to-plasma ratio ranged from 0.6 to 1.2 among the species tested. GDC-0941 human hepatic clearance was predicted to be moderate by liver microsomal incubations. GDC-0941 had high permeability in Madin-Darby canine kidney cells. The clearance of GDC-0941 was high in mouse (63.7 mL/min/kg), rat (49.3 mL/min/kg) and cynomolgus monkey (58.6 mL/min/kg), and moderate in dog (11.9 mL/min/kg). The volume of distribution ranged from 2.52 L/kg in rat to 2.94 L/kg in monkey. Oral bioavailability ranged from 18.6% in monkey to 77.9% in mouse. Predicted human clearance and volume of distribution using allometry were 6 mL/min/kg and 2.9 L/kg, respectively. The human efficacious doses were predicted based on results from preclinical pharmacokinetic studies and xenograft models. GDC-0941 preclinical characterisation and predictions of its properties in human supported its progression towards clinical development. GDC-0941 is currently in phase II clinical trials.


Subject(s)
Indazoles/pharmacokinetics , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacokinetics , Sulfonamides/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Autoradiography , Carbon Radioisotopes , Cell Line , Cell Membrane Permeability , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Humans , Indazoles/administration & dosage , Indazoles/blood , Indazoles/chemistry , Male , Microsomes, Liver/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/chemistry , Species Specificity , Sulfonamides/administration & dosage , Sulfonamides/blood , Sulfonamides/chemistry , Treatment Outcome , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...