Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Drug Metab Dispos ; 44(12): 1881-1889, 2016 12.
Article de Anglais | MEDLINE | ID: mdl-27638506

RÉSUMÉ

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.


Sujet(s)
Tumeurs du cerveau/métabolisme , Encéphale/métabolisme , Glioblastome/métabolisme , Inhibiteurs des phosphoinositide-3 kinases , Inhibiteurs de protéines kinases/métabolisme , Glycoprotéine P/métabolisme , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Animaux , Barrière hémato-encéphalique/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Tumeurs du cerveau/traitement médicamenteux , Lignée cellulaire , Lignée cellulaire tumorale , Chiens , Femelle , Glioblastome/traitement médicamenteux , Gliome/traitement médicamenteux , Gliome/métabolisme , Humains , Indazoles/métabolisme , Indazoles/pharmacologie , Cellules rénales canines Madin-Darby , Souris , Souris nude , Inhibiteurs de protéines kinases/pharmacologie
2.
ACS Med Chem Lett ; 7(4): 351-6, 2016 Apr 14.
Article de Anglais | MEDLINE | ID: mdl-27096040

RÉSUMÉ

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.

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

RÉSUMÉ

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).


Sujet(s)
Antinéoplasiques/pharmacologie , Benzoxépinne/pharmacologie , Conception de médicament , Imidazoles/pharmacologie , Oxazépines/pharmacologie , Inhibiteurs des phosphoinositide-3 kinases , Inhibiteurs de protéines kinases/pharmacologie , Animaux , Antinéoplasiques/composition chimique , Antinéoplasiques/métabolisme , Benzoxépinne/composition chimique , Benzoxépinne/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases de classe I , Chiens , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Humains , Imidazoles/composition chimique , Imidazoles/métabolisme , Isoenzymes/antagonistes et inhibiteurs , Isoenzymes/métabolisme , Macaca fascicularis , Souris , Microsomes du foie/composition chimique , Microsomes du foie/métabolisme , Modèles moléculaires , Structure moléculaire , Oxazépines/composition chimique , Oxazépines/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/métabolisme , Rats , Rat Sprague-Dawley , Relation structure-activité
4.
Nature ; 513(7519): 512-6, 2014 Sep 25.
Article de Anglais | MEDLINE | ID: mdl-25043004

RÉSUMÉ

Mutations that deregulate Notch1 and Ras/phosphoinositide 3 kinase (PI3K)/Akt signalling are prevalent in T-cell acute lymphoblastic leukaemia (T-ALL), and often coexist. Here we show that the PI3K inhibitor GDC-0941 is active against primary T-ALLs from wild-type and Kras(G12D) mice, and addition of the MEK inhibitor PD0325901 increases its efficacy. Mice invariably relapsed after treatment with drug-resistant clones, most of which unexpectedly had reduced levels of activated Notch1 protein, downregulated many Notch1 target genes, and exhibited cross-resistance to γ-secretase inhibitors. Multiple resistant primary T-ALLs that emerged in vivo did not contain somatic Notch1 mutations present in the parental leukaemia. Importantly, resistant clones upregulated PI3K signalling. Consistent with these data, inhibiting Notch1 activated the PI3K pathway, providing a likely mechanism for selection against oncogenic Notch1 signalling. These studies validate PI3K as a therapeutic target in T-ALL and raise the unexpected possibility that dual inhibition of PI3K and Notch1 signalling could promote drug resistance in T-ALL.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Indazoles/pharmacologie , Inhibiteurs des phosphoinositide-3 kinases , Leucémie-lymphome lymphoblastique à précurseurs T/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs T/génétique , Inhibiteurs de protéines kinases/pharmacologie , Récepteur Notch1/métabolisme , Sulfonamides/pharmacologie , Animaux , Benzamides/pharmacologie , Benzamides/usage thérapeutique , Clones cellulaires/effets des médicaments et des substances chimiques , Clones cellulaires/métabolisme , Clones cellulaires/anatomopathologie , Diphénylamine/analogues et dérivés , Diphénylamine/pharmacologie , Diphénylamine/usage thérapeutique , Régulation négative/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Synergie des médicaments , Gènes ras/génétique , Indazoles/usage thérapeutique , Mâle , Souris , Souris de lignée C57BL , Mitogen-Activated Protein Kinase Kinases/antagonistes et inhibiteurs , Leucémie-lymphome lymphoblastique à précurseurs T/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs T/anatomopathologie , Inhibiteurs de protéines kinases/usage thérapeutique , Structure tertiaire des protéines , Protéines proto-oncogènes c-akt/métabolisme , Récepteur Notch1/composition chimique , Récepteur Notch1/déficit , Récepteur Notch1/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Sulfonamides/usage thérapeutique
5.
Mol Cancer Ther ; 12(6): 853-64, 2013 Jun.
Article de Anglais | MEDLINE | ID: mdl-23475955

RÉSUMÉ

Although mitogen-activated protein (MAP)-extracellular signal-regulated kinase (ERK) kinase (MEK) inhibition is predicted to cause cell death by stabilization of the proapoptotic BH3-only protein BIM, the induction of apoptosis is often modest. To determine if addition of a Bcl-2 family inhibitor could increase the efficacy of a MEK inhibitor, we evaluated a panel of 53 non-small cell lung cancer and pancreatic cancer cell lines with the combination of navitoclax (ABT-263), a Bcl-2/Bcl-xL (BCL2/BCL2L1) antagonist, and a novel MAP kinase (MEK) inhibitor, G-963. The combination is synergistic in the majority of lines, with an enrichment of cell lines harboring KRAS mutations in the high synergy group. Cells exposed to G-963 arrest in G1 and a small fraction undergo apoptosis. The addition of navitoclax to G-963 does not alter the kinetics of cell-cycle arrest, but greatly increases the percentage of cells that undergo apoptosis. The G-963/navitoclax combination was more effective than either single agent in the KRAS mutant H2122 xenograft model; BIM stabilization and PARP cleavage were observed in tumors, consistent with the mechanism of action observed in cell culture. Addition of the phosphatidylinositol 3-kinase (PI3K, PIK3CA) inhibitor GDC-0941 to this treatment combination increases cell killing compared with double- or single-agent treatment. Taken together, these data suggest the efficacy of agents that target the MAPK and PI3K pathways can be improved by combination with a Bcl-2 family inhibitor.


Sujet(s)
Tumeurs du poumon/traitement médicamenteux , MAP Kinase Kinase Kinases/antagonistes et inhibiteurs , Tumeurs du pancréas/traitement médicamenteux , Inhibiteurs des phosphoinositide-3 kinases , Protéine bcl-X/antagonistes et inhibiteurs , Dérivés de l'aniline/administration et posologie , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Antienzymes/administration et posologie , Gènes bcl-2/génétique , Humains , Indazoles/administration et posologie , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , MAP Kinase Kinase Kinases/métabolisme , Thérapie moléculaire ciblée , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Sulfonamides/administration et posologie , Protéine bcl-X/génétique
6.
Clin Cancer Res ; 19(7): 1760-72, 2013 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-23287563

RÉSUMÉ

PURPOSE: We describe the preclinical pharmacology and antitumor activity of GDC-0068, a novel highly selective ATP-competitive pan-Akt inhibitor currently in clinical trials for the treatment of human cancers. EXPERIMENTAL DESIGN: The effect of GDC-0068 on Akt signaling was characterized using specific biomarkers of the Akt pathway, and response to GDC-0068 was evaluated in human cancer cell lines and xenograft models with various genetic backgrounds, either as a single agent or in combination with chemotherapeutic agents. RESULTS: GDC-0068 blocked Akt signaling both in cultured human cancer cell lines and in tumor xenograft models as evidenced by dose-dependent decrease in phosphorylation of downstream targets. Inhibition of Akt activity by GDC-0068 resulted in blockade of cell-cycle progression and reduced viability of cancer cell lines. Markers of Akt activation, including high-basal phospho-Akt levels, PTEN loss, and PIK3CA kinase domain mutations, correlate with sensitivity to GDC-0068. Isogenic PTEN knockout also sensitized MCF10A cells to GDC-0068. In multiple tumor xenograft models, oral administration of GDC-0068 resulted in antitumor activity ranging from tumor growth delay to regression. Consistent with the role of Akt in a survival pathway, GDC-0068 also enhanced antitumor activity of classic chemotherapeutic agents. CONCLUSIONS: GDC-0068 is a highly selective, orally bioavailable Akt kinase inhibitor that shows pharmacodynamic inhibition of Akt signaling and robust antitumor activity in human cancer cells in vitro and in vivo. Our preclinical data provide a strong mechanistic rationale to evaluate GDC-0068 in cancers with activated Akt signaling.


Sujet(s)
Antinéoplasiques/pharmacologie , Tumeurs/métabolisme , Pipérazines/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Pyrimidines/pharmacologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Activation enzymatique/effets des médicaments et des substances chimiques , Femelle , Humains , Mâle , Souris , Tumeurs/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
7.
Neoplasia ; 15(12): 1314-29, 2013 Dec.
Article de Anglais | MEDLINE | ID: mdl-24403854

RÉSUMÉ

Nicotinamide adenine dinucleotide (NAD) is a metabolite essential for cell survival and generated de novo from tryptophan or recycled from nicotinamide (NAM) through the nicotinamide phosphoribosyltransferase (NAMPT)-dependent salvage pathway. Alternatively, nicotinic acid (NA) is metabolized to NAD through the nicotinic acid phosphoribosyltransferase domain containing 1 (NAPRT1)-dependent salvage pathway. Tumor cells are more reliant on the NAMPT salvage pathway making this enzyme an attractive therapeutic target. Moreover, the therapeutic index of NAMPT inhibitors may be increased by in NAPRT-deficient tumors by NA supplementation as normal tissues may regenerate NAD through NAPRT1. To confirm the latter, we tested novel NAMPT inhibitors, GNE-617 and GNE-618, in cell culture- and patient-derived tumor models. While NA did not protect NAPRT1-deficient tumor cell lines from NAMPT inhibition in vitro, it rescued efficacy of GNE-617 and GNE-618 in cell culture- and patient-derived tumor xenografts in vivo. NA co-treatment increased NAD and NAM levels in NAPRT1-deficient tumors to levels that sustained growth in vivo. Furthermore, NAM co-administration with GNE-617 led to increased tumor NAD levels and rescued in vivo efficacy as well. Importantly, tumor xenografts remained NAPRT1-deficient in the presence of NA, indicating that the NAPRT1-dependent pathway is not reactivated. Protection of NAPRT1-deficient tumors in vivo may be due to increased circulating levels of metabolites generated by mouse liver, in response to NA or through competitive reactivation of NAMPT by NAM. Our results have important implications for the development of NAMPT inhibitors when considering NA co-treatment as a rescue strategy.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Composés hétérobicycliques/administration et posologie , Pentosyltransferases/déficit , Sulfones/administration et posologie , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lignée cellulaire tumorale , Cytokines/antagonistes et inhibiteurs , Cytokines/génétique , Cytokines/métabolisme , Synergie des médicaments , Femelle , Expression des gènes , Humains , Souris , Souris nude , NAD/métabolisme , Acide nicotinique/administration et posologie , Nicotinamide/administration et posologie , Nicotinamide phosphoribosyltransferase/antagonistes et inhibiteurs , Nicotinamide phosphoribosyltransferase/génétique , Nicotinamide phosphoribosyltransferase/métabolisme , Pentosyltransferases/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Clin Cancer Res ; 18(22): 6239-48, 2012 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-22992516

RÉSUMÉ

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.


Sujet(s)
Antinéoplasiques/pharmacocinétique , Barrière hémato-encéphalique/métabolisme , Tumeurs du cerveau/traitement médicamenteux , Glioblastome/traitement médicamenteux , Inhibiteurs des phosphoinositide-3 kinases , Inhibiteurs de protéines kinases/pharmacocinétique , Pyrimidines/pharmacocinétique , Thiophènes/pharmacocinétique , Animaux , Antinéoplasiques/pharmacologie , Tumeurs du cerveau/enzymologie , Tumeurs du cerveau/anatomopathologie , Perméabilité capillaire , Lignée cellulaire , Perméabilité des membranes cellulaires , Chiens , Femelle , Glioblastome/enzymologie , Glioblastome/anatomopathologie , Humains , Souris , Thérapie moléculaire ciblée , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes c-akt/métabolisme , Pyrimidines/pharmacologie , Thiophènes/pharmacologie , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
9.
J Med Chem ; 55(18): 8007-20, 2012 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-22946614

RÉSUMÉ

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.


Sujet(s)
Barrière hémato-encéphalique/métabolisme , Conception de médicament , Antienzymes/métabolisme , Antienzymes/pharmacologie , Inhibiteurs des phosphoinositide-3 kinases , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Chiens , Antienzymes/composition chimique , Antienzymes/pharmacocinétique , Femelle , Humains , Liaison hydrogène , Cellules rénales canines Madin-Darby , Souris , Perméabilité , Propriétés de surface
10.
Drug Metab Dispos ; 40(9): 1785-96, 2012 Sep.
Article de Anglais | MEDLINE | ID: mdl-22696419

RÉSUMÉ

(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.


Sujet(s)
Antinéoplasiques/pharmacocinétique , Tumeurs du sein/traitement médicamenteux , Composés hétérocycliques bicycliques/pharmacocinétique , Absorption intestinale , Modèles biologiques , Inhibiteurs des phosphoinositide-3 kinases , Inhibiteurs de protéines kinases/pharmacocinétique , Pyrimidines/pharmacocinétique , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Sous-famille B de transporteurs à cassette liant l'ATP/déficit , Sous-famille B de transporteurs à cassette liant l'ATP/génétique , Sous-famille B de transporteurs à cassette liant l'ATP/métabolisme , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP , Transporteurs ABC/déficit , Transporteurs ABC/génétique , Administration par voie orale , Animaux , Antinéoplasiques/administration et posologie , Antinéoplasiques/sang , Aire sous la courbe , Biodisponibilité , Biotransformation , Encéphale/métabolisme , Tumeurs du sein/enzymologie , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Composés hétérocycliques bicycliques/administration et posologie , Composés hétérocycliques bicycliques/sang , Perméabilité des membranes cellulaires , Chiens , Calcul des posologies , Femelle , Période , Hépatocytes/métabolisme , Humains , Injections veineuses , Foie/métabolisme , Cellules MCF-7 , Macaca fascicularis , Cellules rénales canines Madin-Darby , Mâle , Taux de clairance métabolique , Souris , Souris knockout , Phosphatidylinositol 3-kinase/métabolisme , Liaison aux protéines , Inhibiteurs de protéines kinases/administration et posologie , Inhibiteurs de protéines kinases/sang , Pyrimidines/administration et posologie , Pyrimidines/sang , Rats , Rat Sprague-Dawley , Récepteur ErbB-2/génétique , Récepteur ErbB-2/métabolisme , Spécificité d'espèce , Sérine-thréonine kinases TOR/métabolisme , Distribution tissulaire , Transfection , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Clin Cancer Res ; 18(14): 3901-11, 2012 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-22586300

RÉSUMÉ

PURPOSE: Docetaxel is a front-line standard-of-care chemotherapeutic drug for the treatment of breast cancer. Phosphoinositide 3-kinases (PI3K) are lipid kinases that regulate breast tumor cell growth, migration, and survival. The current study was intended to determine whether GDC-0941, an orally bioavailable class I selective PI3K inhibitor, enhances the antitumor activity of docetaxel in human breast cancer models in vitro and in vivo. EXPERIMENTAL DESIGN: A panel of 25 breast tumor cell lines representing HER2+, luminal, and basal subtypes were treated with GDC-0941, docetaxel, or the combination of both drugs and assayed for cellular viability, modulation of PI3K pathway markers, and apoptosis induction. Drug combination effects on cellular viability were also assessed in nontransformed MCF10A human mammary epithelial cells. Human xenografts of breast cancer cell lines and patient-derived tumors were used to assess efficacy of GDC-0941 and docetaxel in vivo. RESULTS: Combination of GDC-0941 and docetaxel decreased the cellular viability of breast tumor cell lines in vitro but to variable degrees of drug synergy. Compared with nontransformed MCF10A cells, the addition of both drugs resulted in stronger synergistic effects in a subset of tumor cell lines that were not predicted by breast cancer subtype. In xenograft models, GDC-0941 enhanced the antitumor activity of docetaxel with maximum combination efficacy observed within 1 hour of administering both drugs. GDC-0941 increased the rate of apoptosis in cells arrested in mitosis upon cotreatment with docetaxel. CONCLUSION: GDC-0941 augments the efficacy of docetaxel by increasing drug-induced apoptosis in breast cancer models.


Sujet(s)
Tumeurs du sein/traitement médicamenteux , Indazoles/administration et posologie , Phosphatidylinositol 3-kinases , Sulfonamides/administration et posologie , Taxoïdes/administration et posologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Docetaxel , Synergie des médicaments , Femelle , Humains , Souris , Tumeurs expérimentales/traitement médicamenteux , Phosphatidylinositol 3-kinases/métabolisme , Inhibiteurs des phosphoinositide-3 kinases
12.
Drug Metab Dispos ; 38(9): 1422-6, 2010 Sep.
Article de Anglais | MEDLINE | ID: mdl-20522663

RÉSUMÉ

2-(1H-Indazol-4-yl)-6-(4-methanesulfonyl-piperazin-1-ylmethyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidine (GDC-0941) is a novel small molecule inhibitor of the phosphatidylinositol 3-kinase (PI3K) pathway currently evaluated in the clinic as an anticancer agent. The objectives of this study were to determine in vitro whether GDC-0941 was a substrate of P-glycoprotein (P-gp) and breast cancer resistance protein (Bcrp1) and to investigate the impact of these transporters on the pharmacokinetics, brain penetration, and activity of GDC-0941 in FVBn mice (wild-type) and Mdr1a/b(-/-), Bcrp1(-/-), and Mdr1a/b(-/-)/Bcrp1(-/-) knockout mice. Studies with Madin-Darby canine kidney cells transfected with P-gp or Bcrp1 established that this compound was a substrate of both transporters. After administrations to mice, GDC-0941 brain-to-plasma ratio ranged from 0.02 to 0.06 in the wild-type and Bcrp1(-/-) mice and was modestly higher in the Mdr1a/b(-/-) mice, ranging from 0.08 to 0.11. In contrast, GDC-0941 brain-to-plasma ratio in Mdr1a/b(-/-)/Bcrp1(-/-) triple knockout mice was 30-fold higher than in the wild-type mice. The plasma clearance of GDC-0941 was similar in wild-type and all knockout mice, ranging from 15 to 25 ml/(min . kg) in the wild-type mice and from 18 to 35 ml/(min . kg) in the knockout mice. Exposure after oral administration was comparable in the four strains of mice. The PI3K pathway was markedly inhibited in the brain of Mdr1a/b(-/-)/Bcrp1(-/-) mice for up to 6 h postdose, as evidenced by a 60% suppression of the phosphorylated Akt signal, whereas no inhibition was detected in the brain of wild-type mice. The concerted effects of P-gp and Bcrp1 in restricting GDC-0941 access and pathway modulation in mouse brain may have implications for the treatment of patients with brain tumors.


Sujet(s)
Glycoprotéine P/physiologie , Transporteurs ABC/physiologie , Encéphale/métabolisme , Indazoles/pharmacologie , Protéines tumorales/physiologie , Inhibiteurs de protéines kinases/pharmacologie , Sulfonamides/pharmacologie , Glycoprotéine P/génétique , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP , Transporteurs ABC/génétique , Animaux , Indazoles/pharmacocinétique , Mâle , Souris , Souris knockout , Protéines tumorales/génétique , Inhibiteurs de protéines kinases/pharmacocinétique , Sulfonamides/pharmacocinétique
13.
Cancer Res ; 70(3): 1164-72, 2010 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-20103642

RÉSUMÉ

Therapeutic inhibitors are being developed against the phosphoinositide 3-kinase (PI3K) pathway, the deregulation of which drives tumor growth and survival in many cancers. There are eight PI3Ks in mammals divided into three classes. Class IA PI3Ks (p110alpha, p110beta, and p110delta) are critical for cell growth and survival, with the p110alpha isoform implicated as the most important in carcinomas. In this study, we examined the effects of small-molecule inhibitors of class IA PI3Ks to explore the contributions of different isoforms in cancer cells. Similar responses were seen in cancer cells with wild-type or activated mutant PI3K genes treated with p110alpha/delta or p110alpha/beta/delta inhibitors in cell viability assays. In contrast, PTEN-negative cell lines tended to be less responsive (4-fold overall) to an inhibitor of p110alpha/delta versus p110alpha/beta/delta. Combining a p110alpha/delta inhibitor with a p110beta inhibitor resulted in comparable potency to the p110alpha/beta/delta inhibitor. The disparity in efficacy was confirmed in vivo. Pharmacodynamic biomarker analysis revealed that an inhibitor with insufficient potency against the p110beta isoform was less effective at inhibiting the PI3K pathway in PTEN-negative tumor xenografts. Our results imply that patients with PTEN-negative tumors may preferentially benefit from treatment with a class I PI3K inhibitor that is capable of inhibiting the p110beta isoform.


Sujet(s)
Inhibiteurs des phosphoinositide-3 kinases , Tumeurs de la prostate/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Animaux , Technique de Western , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Femelle , Cytométrie en flux , Humains , Indazoles/composition chimique , Indazoles/pharmacologie , Isoenzymes/antagonistes et inhibiteurs , Isoenzymes/métabolisme , Mâle , Souris , Souris nude , Structure moléculaire , Mutation , Protéine oncogène v-akt/métabolisme , Phosphohydrolase PTEN/génétique , Phosphohydrolase PTEN/métabolisme , Phosphatidylinositol 3-kinases/génétique , Phosphatidylinositol 3-kinases/métabolisme , Phosphorylation , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Inhibiteurs de protéines kinases/composition chimique , Sulfonamides/composition chimique , Sulfonamides/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
14.
J Exp Med ; 201(6): 925-35, 2005 Mar 21.
Article de Anglais | MEDLINE | ID: mdl-15781583

RÉSUMÉ

Pulmonary fibrosis is the consequence of a variety of diseases with no satisfying treatment option. Therapy-induced fibrosis also limits the efficacy of chemotherapy and radiotherapy in numerous cancers. Here, we studied the potential of platelet-derived growth factor (PDGF) receptor tyrosine kinase inhibitors (RTKIs) to attenuate radiation-induced pulmonary fibrosis. Thoraces of C57BL/6 mice were irradiated (20 Gy), and mice were treated with three distinct PDGF RTKIs (SU9518, SU11657, or Imatinib). Irradiation was found to induce severe lung fibrosis resulting in dramatically reduced mouse survival. Treatment with PDGF RTKIs markedly attenuated the development of pulmonary fibrosis in excellent correlation with clinical, histological, and computed tomography results. Importantly, RTKIs also prolonged the life span of irradiated mice. We found that radiation up-regulated expression of PDGF (A-D) isoforms leading to phosphorylation of PDGF receptor, which was strongly inhibited by RTKIs. Our findings suggest a pivotal role of PDGF signaling in the pathogenesis of pulmonary fibrosis and indicate that inhibition of fibrogenesis, rather than inflammation, is critical to antifibrotic treatment. This study points the way to a potential new approach for treating idiopathic or therapy-related forms of lung fibrosis.


Sujet(s)
Rayons gamma/effets indésirables , Facteur de croissance dérivé des plaquettes/métabolisme , Inhibiteurs de protéines kinases/administration et posologie , Fibrose pulmonaire/traitement médicamenteux , Récepteurs aux facteurs de croissance dérivés des plaquettes/antagonistes et inhibiteurs , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Femelle , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des radiations , Humains , Inflammation/traitement médicamenteux , Inflammation/physiopathologie , Souris , Isoformes de protéines/métabolisme , Fibrose pulmonaire/étiologie , Fibrose pulmonaire/physiopathologie , Transduction du signal/effets des radiations , Irradiation corporelle totale
15.
Clin Cancer Res ; 9(15): 5729-34, 2003 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-14654558

RÉSUMÉ

PURPOSE: The purpose of this study was to evaluate the effect of the receptor tyrosine kinase inhibitor SU11654 on the activity of its molecular target KIT in canine mast cell tumors (MCT) and correlate target inhibition with mutational status of the c-kit juxtamembrane domain and SU11654 plasma concentration. EXPERIMENTAL DESIGN: Tumor biopsies were obtained from dogs with advanced MCTs before and 8 h after administration of a single oral dose of SU11654, previously shown to be active in dogs with MCTs. Blood samples were taken to determine the plasma concentration of SU11654. Levels of phosphorylated KIT and ERK1/2 were assessed in tumor biopsies by Western blot. Tumors were analyzed by PCR for the presence or absence of an internal tandem duplication (ITD) in the juxtamembrane domain of c-kit. RESULTS: Fourteen dogs with advanced MCTs were enrolled in the study; 11 of these were evaluable for KIT target modulation (the remaining tumor specimens had inevaluable amounts of total KIT protein). Of these, eight MCTs showed reduced levels of phosphorylated KIT relative to total KIT after treatment with SU11654, compared with pretreatment biopsies. All four evaluable MCTs expressing ITD mutant c-kitshowed modulation of KIT phosphorylation, as did four of seven tumors expressing non-ITD c-kit. Phosphorylated ERK1/2 was modulated in seven tumors; this did not correlate with inhibition of KIT phosphorylation CONCLUSION: SU11654 treatment at the efficacious dose results in inhibition of KIT phosphorylation in canine MCTs.


Sujet(s)
Maladies des chiens/traitement médicamenteux , Antienzymes/usage thérapeutique , Indoles/usage thérapeutique , Sarcome à mastocytes/médecine vétérinaire , Protéines proto-oncogènes c-kit/métabolisme , Pyrroles/usage thérapeutique , Animaux , Maladies des chiens/anatomopathologie , Chiens , Antienzymes/sang , Antienzymes/pharmacocinétique , Indoles/sang , Indoles/pharmacocinétique , Mastocytes , Sarcome à mastocytes/traitement médicamenteux , Sarcome à mastocytes/anatomopathologie , Mitogen-Activated Protein Kinases/métabolisme , Métastase tumorale , Phosphorylation , Pyrroles/sang , Pyrroles/pharmacocinétique , Récidive
16.
Mol Cancer Ther ; 2(10): 1011-21, 2003 Oct.
Article de Anglais | MEDLINE | ID: mdl-14578466

RÉSUMÉ

SU11248 is an oral multitargeted tyrosine kinase inhibitor with antitumor and antiangiogenic activities through targeting platelet-derived growth factor receptor, vascular endothelial growth factor receptor, KIT, and FLT3, the first three of which are expressed in human breast cancer and/or its supporting tissues. The purpose of the present studies was to demonstrate the potent anticancer activity of SU11248 alone or in combination with conventional cytotoxic agents against several distinct preclinical models of breast cancer. SU11248 was administered as a monotherapy to (1) mouse mammary tumor virus-v-Ha-ras mice and 7,12-dimethylbenz(a)anthracene-treated rats bearing mammary tumors and (2) mice bearing human breast cancer xenografts of s.c. MX-1 tumors and osseous metastasis of a MDA-MB-435-derived cell line (435/HAL-Luc). SU11248 was also administered in combination with docetaxel both in xenograft models and in combination with 5-fluorouracil and doxorubicin in the MX-1 model. SU11248 treatment potently regressed growth of mammary cancers in mouse mammary tumor virus-v-Ha-ras transgenic mice (82% regression) and 7,12-dimethylbenz(a)anthracene-induced mammary tumors in rats (99% regression at the highest dose; P < 0.05 for both). This agent also inhibited MX-1 tumor growth by 52%, with markedly enhanced anticancer effects when administered in combination with docetaxel, 5-fluorouracil, or doxorubicin compared with either agent alone (P < 0.05). SU11248 treatment in combination with docetaxel effectively prolonged survival of mice, with 435/HAL-Luc cancer xenografts established in bone compared with either agent alone (P < 0.05). These results demonstrate that SU11248 is effective in preclinical breast cancer models and suggest that it may be useful in the treatment of breast cancer in the clinic.


Sujet(s)
Tumeurs du sein/traitement médicamenteux , Antienzymes/pharmacologie , Indoles/pharmacologie , Pyrroles/pharmacologie , Animaux , Antibiotiques antinéoplasiques/pharmacologie , Antibiotiques antinéoplasiques/usage thérapeutique , Tumeurs osseuses/secondaire , Lignée cellulaire tumorale , Doxorubicine/pharmacologie , Doxorubicine/usage thérapeutique , Femelle , Fluorouracil/usage thérapeutique , Humains , Souris , Souris nude , Souris transgéniques , Transplantation tumorale , Phosphorylation , Rats , Rat Sprague-Dawley , Sunitinib , Facteurs temps
17.
Mol Cancer Ther ; 2(5): 471-8, 2003 May.
Article de Anglais | MEDLINE | ID: mdl-12748309

RÉSUMÉ

The purpose of this study was to evaluate the activity of the indolinone kinase inhibitor SU11248 against the receptor tyrosine kinase KIT in vitro and in vivo, examine the role of KIT in small cell lung cancer (SCLC), and anticipate clinical utility of SU11248 in SCLC. SU11248 is an oral, multitargeted tyrosine kinase inhibitor with direct antitumor and antiangiogenic activity through targeting platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor receptor, KIT, and FLT3 receptors. Treatment of the KIT-expressing SCLC-derived NCI-H526 cell line in vitro with SU11248 resulted in dose-dependent inhibition of stem cell factor-stimulated KIT phosphotyrosine levels and proliferation. The biological significance of KIT inhibition was evaluated in vivo by treating mice bearing s.c. NCI-H526 tumors with SU11248 or another structurally unrelated KIT inhibitor, STI571 (Gleevec), which is also known to inhibit Bcr-Abl and PDGFRbeta. SU11248 treatment resulted in significant tumor growth inhibition, whereas inhibition from STI571 treatment was less dramatic. Both compounds reduced phospho-KIT levels in NCI-H526 tumors, with a greater reduction by SU11248, correlating with efficacy. Likewise, phospho-PDGFRbeta levels contributed by tumor stroma and with known involvement in angiogenesis were strongly inhibited by SU11248 and less so by STI571. Because platinum-based chemotherapy is part of the standard of care for SCLC, SU11248 was combined with cisplatin, and significant tumor growth delay was measured compared with either agent alone. These results expand the profile of SU11248 as a KIT signaling inhibitor and suggest that SU11248 may have clinical potential in the treatment of SCLC via direct antitumor activity mediated via KIT as well as tumor angiogenesis via vascular endothelial growth factor receptor FLK1/KDR and PDGFRbeta.


Sujet(s)
Carcinome à petites cellules/métabolisme , Indoles/pharmacologie , Tumeurs du poumon/métabolisme , Protéines proto-oncogènes c-kit/effets des médicaments et des substances chimiques , Pyrroles/pharmacologie , Récepteur au PDGF bêta/antagonistes et inhibiteurs , Animaux , Antinéoplasiques/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Benzamides , Carcinome à petites cellules/anatomopathologie , Division cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Évaluation préclinique de médicament , Antienzymes/pharmacologie , Femelle , Humains , Mésilate d'imatinib , Tumeurs du poumon/anatomopathologie , Souris , Souris nude , Phosphorylation , Phosphotyrosine/métabolisme , Pipérazines/pharmacologie , Protéines proto-oncogènes c-kit/métabolisme , Pyrimidines/pharmacologie , Récepteur au PDGF bêta/métabolisme , Facteur de croissance des cellules souches/physiologie , Sunitinib , Cellules cancéreuses en culture/transplantation
18.
Blood ; 101(9): 3597-605, 2003 May 01.
Article de Anglais | MEDLINE | ID: mdl-12531805

RÉSUMÉ

FLT3 (fms-related tyrosine kinase/Flk2/Stk-2) is a receptor tyrosine kinase (RTK) primarily expressed on hematopoietic cells. In blasts from acute myelogenous leukemia (AML) patients, 2 classes of FLT3 activating mutations have been identified: internal tandem duplication (ITD) mutations in the juxtamembrane domain (25%-30% of patients) and point mutations in the kinase domain activation loop (7%-8% of patients). FLT3-ITD mutations are the most common molecular defect identified in AML and have been shown to be an independent prognostic factor for decreased survival. FLT3-ITD is therefore an attractive molecular target for therapy. SU11248 is a recently described selective inhibitor with selectivity for split kinase domain RTKs, including platelet-derived growth factor receptors, vascular endothelial growth factor receptors, and KIT. We show that SU11248 also has potent activity against wild-type FLT3 (FLT3-WT), FLT3-ITD, and FLT3 activation loop (FLT3-Asp835) mutants in phosphorylation assays. SU11248 inhibits FLT3-driven phosphorylation and induces apoptosis in vitro. In addition, SU11248 inhibits FLT3-induced VEGF production. The in vivo efficacy of SU11248 was investigated in 2 FLT3-ITD models: a subcutaneous tumor xenograft model and a bone marrow engraftment model. We show that SU11248 (20 mg/kg/d) dramatically regresses FLT3-ITD tumors in the subcutaneous tumor xenograft model and prolongs survival in the bone marrow engraftment model. Pharmacokinetic and pharmacodynamic analysis in subcutaneous tumors showed that a single administration of an efficacious drug dose potently inhibits FLT3-ITD phosphorylation for up to 16 hours following a single dose. These results suggest that further exploration of SU11248 activity in AML patients is warranted.


Sujet(s)
Antinéoplasiques/pharmacologie , Antienzymes/pharmacologie , Indoles/pharmacologie , Protéines proto-oncogènes/antagonistes et inhibiteurs , Pyrroles/pharmacologie , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Maladie aigüe , Substitution d'acide aminé , Animaux , Antinéoplasiques/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Transplantation de moelle osseuse , Facteurs de croissance endothéliale/biosynthèse , Antienzymes/usage thérapeutique , Femelle , Humains , Indoles/usage thérapeutique , Protéines et peptides de signalisation intercellulaire/biosynthèse , Leucémie myéloïde/enzymologie , Leucémie myéloïde/anatomopathologie , Lymphokines/biosynthèse , Souris , Souris de lignée NOD , Souris nude , Souris SCID , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/génétique , Phosphorylation/effets des médicaments et des substances chimiques , Mutation ponctuelle , Maturation post-traductionnelle des protéines/effets des médicaments et des substances chimiques , Structure tertiaire des protéines , Protéines proto-oncogènes/génétique , Pyrroles/usage thérapeutique , Récepteurs à activité tyrosine kinase/génétique , Protéines de fusion recombinantes/antagonistes et inhibiteurs , Transduction du signal/effets des médicaments et des substances chimiques , Sunitinib , Séquences répétées en tandem , Transfection , Cellules cancéreuses en culture/enzymologie , Facteur de croissance endothéliale vasculaire de type A , Facteurs de croissance endothéliale vasculaire , Tests d'activité antitumorale sur modèle de xénogreffe , Tyrosine kinase-3 de type fms
19.
Clin Cancer Res ; 9(1): 327-37, 2003 Jan.
Article de Anglais | MEDLINE | ID: mdl-12538485

RÉSUMÉ

One challenging aspect in the clinical development of molecularly targeted therapies, which represent a new and promising approach to treating cancers, has been the identification of a biologically active dose rather than a maximum tolerated dose. The goal of the present study was to identify a pharmacokinetic/pharmacodynamic relationship in preclinical models that could be used to help guide selection of a clinical dose. SU11248, a novel small molecule receptor tyrosine kinase inhibitor with direct antitumor as well as antiangiogenic activity via targeting the vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), KIT, and FLT3 receptor tyrosine kinases, was used as the pharmacological agent in these studies. In mouse xenograft models, SU11248 exhibited broad and potent antitumor activity causing regression, growth arrest, or substantially reduced growth of various established xenografts derived from human or rat tumor cell lines. To predict the target SU11248 exposure required to achieve antitumor activity in mouse xenograft models, we directly measured target phosphorylation in tumor xenografts before and after SU11248 treatment and correlated this with plasma inhibitor levels. In target modulation studies in vivo, SU11248 selectively inhibited Flk-1/KDR (VEGF receptor 2) and PDGF receptor beta phosphorylation (in a time- and dose-dependent manner) when plasma concentrations of inhibitor reached or exceeded 50-100 ng/ml. Similar results were obtained in a functional assay of VEGF-induced vascular permeability in vivo. Constant inhibition of VEGFR2 and PDGF receptor beta phosphorylation was not required for efficacy; at highly efficacious doses, inhibition was sustained for 12 h of a 24-h dosing interval. The pharmacokinetic/pharmacodynamic relationship established for SU11248 in these preclinical studies has aided in the design, selection, and evaluation of dosing regimens being tested in human trials.


Sujet(s)
Antinéoplasiques/pharmacologie , Antienzymes/pharmacologie , Indoles/pharmacologie , Protein-tyrosine kinases/antagonistes et inhibiteurs , Pyrroles/pharmacologie , Récepteurs aux facteurs de croissance dérivés des plaquettes/métabolisme , Récepteurs aux facteurs de croissance endothéliale vasculaire/métabolisme , Animaux , Division cellulaire/effets des médicaments et des substances chimiques , Femelle , Humains , Concentration inhibitrice 50 , Cinétique , Souris , Souris nude , Modèles chimiques , Transplantation tumorale , Phosphorylation , Sunitinib , Facteurs temps , Cellules cancéreuses en culture
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE