Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 14 de 14
Filtrer
2.
Nat Cancer ; 5(7): 1102-1120, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38565920

RÉSUMÉ

The YAP-TEAD protein-protein interaction mediates YAP oncogenic functions downstream of the Hippo pathway. To date, available YAP-TEAD pharmacologic agents bind into the lipid pocket of TEAD, targeting the interaction indirectly via allosteric changes. However, the consequences of a direct pharmacological disruption of the interface between YAP and TEADs remain largely unexplored. Here, we present IAG933 and its analogs as potent first-in-class and selective disruptors of the YAP-TEAD protein-protein interaction with suitable properties to enter clinical trials. Pharmacologic abrogation of the interaction with all four TEAD paralogs resulted in YAP eviction from chromatin and reduced Hippo-mediated transcription and induction of cell death. In vivo, deep tumor regression was observed in Hippo-driven mesothelioma xenografts at tolerated doses in animal models as well as in Hippo-altered cancer models outside mesothelioma. Importantly this also extended to larger tumor indications, such as lung, pancreatic and colorectal cancer, in combination with RTK, KRAS-mutant selective and MAPK inhibitors, leading to more efficacious and durable responses. Clinical evaluation of IAG933 is underway.


Sujet(s)
Voie de signalisation Hippo , Protein-Serine-Threonine Kinases , Facteurs de transcription , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Animaux , Facteurs de transcription/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Souris , Lignée cellulaire tumorale , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines de signalisation YAP/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Protéines de liaison à l'ADN/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs de transcription à domaine TEA , Protéines G ras/métabolisme , Femelle , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique
3.
Cancer Res ; 82(6): 1098-1109, 2022 03 15.
Article de Anglais | MEDLINE | ID: mdl-35131871

RÉSUMÉ

Preventing development of childhood B-cell acute lymphoblastic leukemia (B-ALL), a disease with devastating effects, is a longstanding and unsolved challenge. Heterozygous germline alterations in the PAX5 gene can lead to B-ALL upon accumulation of secondary mutations affecting the JAK/STAT signaling pathway. Preclinical studies have shown that this malignant transformation occurs only under immune stress such as exposure to infectious pathogens. Here we show in Pax5+/- mice that transient, early-life administration of clinically relevant doses of ruxolitinib, a JAK1/2 inhibitor, significantly mitigates the risk of B-ALL following exposure to infection; 1 of 29 animals treated with ruxolitinib developed B-ALL versus 8 of 34 untreated mice. Ruxolitinib treatment preferentially targeted Pax5+/- versus wild-type B-cell progenitors and exerted unique effects on the Pax5+/- B-cell progenitor transcriptional program. These findings provide the first in vivo evidence for a potential strategy to prevent B-ALL development. SIGNIFICANCE: JAK/STAT inhibition suppresses tumorigenesis in a B-ALL-susceptible mouse model, presenting a novel approach to prevent B-ALL onset.


Sujet(s)
Janus kinases , Leucémie-lymphome lymphoblastique à précurseurs B et T , Animaux , Humains , Janus kinases/génétique , Souris , Protéine activatrice spécifique des lymphocytes B/génétique , Facteurs de transcription STAT , Transduction du signal/génétique
4.
Front Pharmacol ; 12: 785851, 2021.
Article de Anglais | MEDLINE | ID: mdl-35342386

RÉSUMÉ

Understanding the pharmacokinetic/pharmacodynamic (PK/PD)-relationship of a drug candidate is key to determine effective, yet safe treatment regimens for patients. However, current testing strategies are inefficient in characterizing in vivo responses to fluctuating drug concentrations during multi-day treatment cycles. Methods based on animal models are resource-intensive and require time, while traditional in vitro cell-culturing methods usually do not provide temporally-resolved information on the effects of in vivo-like drug exposure scenarios. To address this issue, we developed a microfluidic system to 1) culture arrays of three-dimensional spheroids in vitro, to 2) apply specific dynamic drug exposure profiles, and to 3) in-situ analyze spheroid growth and the invoked drug effects in 3D by means of 2-photon microscopy at tissue and single-cell level. Spheroids of fluorescently-labeled T-47D breast cancer cells were monitored under perfusion-culture conditions at short time intervals over three days and exposed to either three 24 h-PK-cycles or a dose-matched constant concentration of the phosphatidylinositol 3-kinase inhibitor BYL719. While the overall efficacy of the two treatment regimens was similar, spheroids exposed to the PK profile displayed cycle-dependent oscillations between regression and regrowth. Spheroids treated with a constant BYL719 concentration regressed at a steady, albeit slower rate. At a single-cell level, the cell density in BYL719-treated spheroids oscillated in a concentration-dependent manner. Our system represents a versatile tool for in-depth preclinical characterization of PK/PD parameters, as it enables an evaluation of drug efficacy and/or toxicity under realistic exposure conditions.

5.
J Med Chem ; 63(21): 12542-12573, 2020 11 12.
Article de Anglais | MEDLINE | ID: mdl-32930584

RÉSUMÉ

FGF19 signaling through the FGFR4/ß-klotho receptor complex has been shown to be a key driver of growth and survival in a subset of hepatocellular carcinomas, making selective FGFR4 inhibition an attractive treatment opportunity. A kinome-wide sequence alignment highlighted a poorly conserved cysteine residue within the FGFR4 ATP-binding site at position 552, two positions beyond the gate-keeper residue. Several strategies for targeting this cysteine to identify FGFR4 selective inhibitor starting points are summarized which made use of both rational and unbiased screening approaches. The optimization of a 2-formylquinoline amide hit series is described in which the aldehyde makes a hemithioacetal reversible-covalent interaction with cysteine 552. Key challenges addressed during the optimization are improving the FGFR4 potency, metabolic stability, and solubility leading ultimately to the highly selective first-in-class clinical candidate roblitinib.


Sujet(s)
Pipérazines/composition chimique , Inhibiteurs de protéines kinases/composition chimique , Pyridines/composition chimique , Récepteur FGFR4/antagonistes et inhibiteurs , Séquence d'acides aminés , Animaux , Sites de fixation , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cystéine/composition chimique , Chiens , Conception de médicament , Période , Hépatocytes/cytologie , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Tumeurs du foie/traitement médicamenteux , Souris , Microsomes du foie/métabolisme , Simulation de dynamique moléculaire , Pipérazines/métabolisme , Pipérazines/pharmacologie , Pipérazines/usage thérapeutique , Inhibiteurs de protéines kinases/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Pyridines/métabolisme , Pyridines/pharmacologie , Pyridines/usage thérapeutique , Rats , Récepteur FGFR4/métabolisme , Relation structure-activité , Tests d'activité antitumorale sur modèle de xénogreffe
6.
Mol Cancer Ther ; 18(12): 2194-2206, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31409633

RÉSUMÉ

Hepatocellular carcinoma (HCC) is the most common primary malignancy of the liver and it is the third leading cause of cancer-related deaths worldwide. Recently, aberrant signaling through the FGF19/FGFR4 axis has been implicated in HCC. Here, we describe the development of FGF401, a highly potent and selective, first in class, reversible-covalent small-molecule inhibitor of the kinase activity of FGFR4. FGF401 is exquisitely selective for FGFR4 versus the other FGFR paralogues FGFR1, FGFR2, FGFR3, and all other kinases in the kinome. FGF401 has excellent drug-like properties showing a robust pharmacokinetic/pharmacodynamics/efficacy relationship, driven by a fraction of time above the phospho-FGFR4 IC90 value. FGF401 has remarkable antitumor activity in mice bearing HCC tumor xenografts and patient-derived xenograft models that are positive for FGF19, FGFR4, and KLB. FGF401 is the first FGFR4 inhibitor to enter clinical trials, and a phase I/II study is currently ongoing in HCC and other solid malignancies.


Sujet(s)
Facteurs de croissance fibroblastique/génétique , Tumeurs du foie/génétique , Tumeurs du foie/thérapie , Récepteur FGFR4/antagonistes et inhibiteurs , Animaux , Humains , Tumeurs du foie/anatomopathologie , Souris , Souris nude , Transduction du signal
7.
Cancer Res ; 78(21): 6257-6267, 2018 11 01.
Article de Anglais | MEDLINE | ID: mdl-30135191

RÉSUMÉ

Activation of p53 by inhibitors of the p53-MDM2 interaction is being pursued as a therapeutic strategy in p53 wild-type cancers. Here, we report distinct mechanisms by which the novel, potent, and selective inhibitor of the p53-MDM2 interaction HDM201 elicits therapeutic efficacy when applied at various doses and schedules. Continuous exposure of HDM201 led to induction of p21 and delayed accumulation of apoptotic cells. By comparison, high-dose pulses of HDM201 were associated with marked induction of PUMA and a rapid onset of apoptosis. shRNA screens identified PUMA as a mediator of the p53 response specifically in the pulsed regimen. Consistent with this, the single high-dose HDM201 regimen resulted in rapid and marked induction of PUMA expression and apoptosis together with downregulation of Bcl-xL in vivo Knockdown of Bcl-xL was identified as the top sensitizer to HDM201 in vitro, and Bcl-xL was enriched in relapsing tumors from mice treated with intermittent high doses of HDM201. These findings define a regimen-dependent mechanism by which disruption of MDM2-p53 elicits therapeutic efficacy when given with infrequent dosing. In an ongoing HDM201 trial, the observed exposure-response relationship indicates that the molecular mechanism elicited by pulse dosing is likely reproducible in patients. These data support the clinical comparison of daily and intermittent regimens of p53-MDM2 inhibitors.Significance: Pulsed high doses versus sustained low doses of the p53-MDM2 inhibitor HDM201 elicit a proapoptotic response from wild-type p53 cancer cells, offering guidance to current clinical trials with this and other drugs that exploit the activity of p53. Cancer Res; 78(21); 6257-67. ©2018 AACR.


Sujet(s)
Antinéoplasiques/administration et posologie , Imidazoles/administration et posologie , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Pyrimidines/administration et posologie , Pyrroles/administration et posologie , Protéine p53 suppresseur de tumeur/antagonistes et inhibiteurs , Animaux , Antinéoplasiques/pharmacologie , Apoptose , Aire sous la courbe , Lignée cellulaire tumorale , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Tests de criblage d'agents antitumoraux , Humains , Imidazoles/pharmacologie , Estimation de Kaplan-Meier , Dose maximale tolérée , Souris , Transplantation tumorale , Pyrimidines/pharmacologie , Pyrroles/pharmacologie , Petit ARN interférent/métabolisme , Facteurs temps , Protéine bcl-X/métabolisme
8.
Anticancer Drugs ; 29(7): 691-701, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-29734209

RÉSUMÉ

Patupilone is a microtubule-targeted cytotoxic agent with clinical efficacy, but causes diarrhoea in more than 80% of patients. The efficacy and tolerability of patupilone delivered continuously by subcutaneous (s.c.) mini-pumps [(mini-pump dose (MPD)] or by intravenous bolus administration [intravenous bolus dose (IVBD)] were compared preclinically to determine whether the therapeutic index could be improved. The antiproliferative potency in vitro of patupilone was determined by measuring total cell protein. Tumours were grown s.c. in rats (A15) or nude mice (KB31, KB8511) or intracranially in nude mice (NCI-H460-Luc). Efficacy was monitored by measuring tumour volumes, bioluminescence or survival. Toxicity was monitored by body weight and/or diarrhoea. Total drug levels in blood, plasma, tissues or dialysates were quantified ex-vivo by liquid chromatography-mass spectroscopy/mass spectroscopy. Patupilone was potent in vitro with GI50s of 0.24-0.28 nmol/l and GI90s of 0.46-1.64 nmol/l. In rats, a single IVBD of patupilone dose dependently inhibited the growth of A15 tumours, but also caused dose-dependent body weight loss and diarrhoea, whereas MPD achieved similar efficacy, but no toxicity. In mice, MPD showed efficacy similar to that of IVBD against KB31 and KB8511 tumours, but with reduced toxicity. In a mouse intracranial tumour model, IVBD was more efficacious than MPD, consistent with patupilone concentrations in the brain. MPD provided constant plasma levels, whereas IVBD had very high C0/Cmin ratios of 70-280 (rat) or 8000 (mouse) over the dosing cycle. Overall, the correlation of plasma and tumour levels with response indicated that a Cave of at least GI90 led to tumour stasis. Continuous low concentrations of patupilone by MPD increased the therapeutic index in s.c. rodent tumour models compared with IVBD by maintaining efficacy, but reducing toxicity.


Sujet(s)
Antinéoplasiques/administration et posologie , Tumeurs du cerveau/traitement médicamenteux , Épothilones/administration et posologie , Pompes à perfusion , Tumeurs expérimentales/traitement médicamenteux , Animaux , Antinéoplasiques/sang , Antinéoplasiques/usage thérapeutique , Tumeurs du cerveau/sang , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Épothilones/sang , Épothilones/usage thérapeutique , Femelle , Humains , Perfusions sous-cutanées , Injections veineuses , Souris , Souris nude , Microdialyse , Tumeurs expérimentales/sang , Tumeurs expérimentales/anatomopathologie , Rats , Spécificité d'espèce , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Cancer Chemother Pharmacol ; 80(4): 869-878, 2017 Oct.
Article de Anglais | MEDLINE | ID: mdl-28779265

RÉSUMÉ

PURPOSE: Everolimus is a drug used successfully in a number of different oncology indications, but significant on-target toxicities exist. We explored the possibility of improving the therapeutic index (TI) by studying alternative means of administering the drug based upon low continuous dosing. METHODS: All studies were performed using naïve nude mice or nude mice bearing s.c. human renal 786-O tumours or human breast MDA-MB-468 tumours. Everolimus was administered via a standard emulsion, either i.v., p.o., i.p., s.c., or via s.c. osmotic mini-pumps (MP) or via poly-lactic-co-glycolic (PLGA)-microparticles (PLGA-µP) prepared from everolimus powder injected s.c. Total-drug levels in blood, plasma or tissues were quantified ex vivo by LC-MS/MS. Efficacy studies were performed over 2-3 weeks and toxicity assessed by changes in body weight, glucose and white blood cell count. Effects on tumour activity biomarkers were quantified using reverse-phase protein array. RESULTS: Everolimus administration s.c. in an emulsion decreased the absorption rate but increased the C max and bio-availability of everolimus compared to standard approaches of administration p.o. or i.p. Everolimus administration s.c. via MP or PLGA-µP reduced the C max and provided continuous low concentrations of everolimus in the plasma, which inhibited tumour pS6/S6 to a similar degree to oral administration. Toxicities such as changes in body weight or white blood cell count were unaffected. Provided the everolimus concentration was above the free unbound IC50 for proliferation of the tumour cell line, efficacy could be achieved equivalent to that provided by standard oral administration. However, an overall improvement in the TI could not be demonstrated. CONCLUSIONS: Continuous low plasma concentrations of everolimus can provide strong efficacy in preclinical models, which if translatable to the clinic may reduce on-target toxicities and so increase the TI.


Sujet(s)
Antinéoplasiques/administration et posologie , Tumeurs du sein/traitement médicamenteux , Évérolimus/administration et posologie , Tumeurs du rein/traitement médicamenteux , Administration par voie orale , Animaux , Antinéoplasiques/pharmacocinétique , Antinéoplasiques/toxicité , Biodisponibilité , Chromatographie en phase liquide/méthodes , Calendrier d'administration des médicaments , Évérolimus/pharmacocinétique , Évérolimus/toxicité , Femelle , Humains , Concentration inhibitrice 50 , Acide lactique/composition chimique , Souris , Souris nude , Acide polyglycolique/composition chimique , Copolymère d'acide poly(lactique-co-glycolique) , Spectrométrie de masse en tandem/méthodes , Tests d'activité antitumorale sur modèle de xénogreffe
10.
Cancer Cell ; 28(1): 29-41, 2015 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-26175414

RÉSUMÉ

A variety of cancers depend on JAK2 signaling, including the high-risk subset of B cell acute lymphoblastic leukemias (B-ALLs) with CRLF2 rearrangements. Type I JAK2 inhibitors induce paradoxical JAK2 hyperphosphorylation in these leukemias and have limited activity. To improve the efficacy of JAK2 inhibition in B-ALL, we developed the type II inhibitor CHZ868, which stabilizes JAK2 in an inactive conformation. CHZ868 potently suppressed the growth of CRLF2-rearranged human B-ALL cells, abrogated JAK2 signaling, and improved survival in mice with human or murine B-ALL. CHZ868 and dexamethasone synergistically induced apoptosis in JAK2-dependent B-ALLs and further improved in vivo survival compared to CHZ868 alone. These data support the testing of type II JAK2 inhibition in patients with JAK2-dependent leukemias and other disorders.


Sujet(s)
Aminopyridines/administration et posologie , Antinéoplasiques/administration et posologie , Benzimidazoles/administration et posologie , Dexaméthasone/administration et posologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Kinase Janus-2/antagonistes et inhibiteurs , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Inhibiteurs de protéines kinases/administration et posologie , Aminopyridines/pharmacologie , Animaux , Antinéoplasiques/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Apoptose , Benzimidazoles/pharmacologie , Lignée cellulaire tumorale , Cytoprotection/effets des médicaments et des substances chimiques , Synergie des médicaments , Humains , Kinase Janus-2/composition chimique , Kinase Janus-2/génétique , Souris , Mutation , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Inhibiteurs de protéines kinases/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Clin Cancer Res ; 19(22): 6230-41, 2013 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-24081976

RÉSUMÉ

PURPOSE: The myeloproliferative neoplasm myelofibrosis is characterized by frequent deregulation of Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling, and JAK inhibitors were shown to reduce splenomegaly and ameliorate disease-related symptoms. However, the mutant clone and bone marrow fibrosis persist in the majority of patients. Using preclinical models, we explored whether JAK and pan-deacetylase inhibitor combination yielded additional benefits. EXPERIMENTAL DESIGN: The combination of the JAK1/2 inhibitor ruxolitinib and panobinostat was investigated using two different mouse models of JAK2(V617F)-driven disease. A Ba/F3 JAK2(V617F) cell-driven leukemic disease model was used to identify tolerated and efficacious doses. The drugs were then evaluated alone and in combination in a mouse model of myeloproliferative neoplasm-like disease based on transplantation of bone marrow transduced with a retrovirus expressing JAK2(V617F). Exposures were determined in blood and tissues, and phosphorylated STAT5 and acetylated histone H3 pharmacodynamic readouts were assessed in spleen and bone marrow. Histologic analysis was conducted on spleen and bone marrow, including staining of reticulin fibers in the latter organ. RESULTS: The combination of ruxolitinib and panobinostat was found to have a more profound effect on splenomegaly, as well as on bone marrow and spleen histology, compared with either agent alone, and the analysis of pharmacodynamic readouts showed that ruxolitinib and panobinostat have nonoverlapping and complementary effects. CONCLUSION: Combining JAK1/2 and pan-deacetylase inhibitors was fairly well tolerated and resulted in improved efficacy in mouse models of JAK2(V617F)-driven disease compared with the single agents. Thus, the combination of ruxolitinib and panobinostat may represent a promising novel therapeutic modality for myeloproliferative neoplasms.


Sujet(s)
Acides hydroxamiques/usage thérapeutique , Indoles/usage thérapeutique , Janus kinase 1/antagonistes et inhibiteurs , Kinase Janus-2/antagonistes et inhibiteurs , Myélofibrose primitive/traitement médicamenteux , Pyrazoles/usage thérapeutique , Acétylation , Animaux , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Cellules de la moelle osseuse/cytologie , Cellules de la moelle osseuse/métabolisme , Modèles animaux de maladie humaine , Inhibiteurs de désacétylase d'histone/effets indésirables , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Histone deacetylases/effets des médicaments et des substances chimiques , Histone/métabolisme , Acides hydroxamiques/effets indésirables , Indoles/effets indésirables , Janus kinase 1/métabolisme , Kinase Janus-2/génétique , Kinase Janus-2/métabolisme , Souris , Nitriles , Panobinostat , Polyglobulie primitive essentielle/traitement médicamenteux , Pyrazoles/effets indésirables , Pyrimidines , Réticuline/analyse , Facteur de transcription STAT-5/effets des médicaments et des substances chimiques , Facteur de transcription STAT-5/métabolisme , Rate/cytologie , Rate/métabolisme , Splénomégalie/traitement médicamenteux , Thrombocytose/traitement médicamenteux
12.
PLoS One ; 7(8): e44146, 2012.
Article de Anglais | MEDLINE | ID: mdl-22952903

RÉSUMÉ

Activating K-RAS mutations occur at a frequency of 90% in pancreatic cancer, and to date no therapies exist targeting this oncogene. K-RAS signals via downstream effector pathways such as the MAPK and the PI3K signaling pathways, and much effort has been focused on developing drugs targeting components of these pathways. To better understand the requirements for K-RAS and its downstream signaling pathways MAPK and PI3K in pancreatic tumor maintenance, we established an inducible K-RAS knock down system that allowed us to ablate K-RAS in established tumors. Knock down of K-RAS resulted in impaired tumor growth in all pancreatic xenograft models tested, demonstrating that K-RAS expression is indeed required for tumor maintenance of K-RAS mutant pancreatic tumors. We further examined signaling downstream of K-RAS, and detected a robust reduction of pERK levels upon K-RAS knock down. In contrast, no effect on pAKT levels could be observed due to almost undetectable basal expression levels. To investigate the requirement of the MAPK and the PI3K pathways on tumor maintenance, three selected pancreatic xenograft models were tested for their response to MEK or PI3K inhibition. Tumors of all three models regressed upon MEK inhibition, but showed less pronounced response to PI3K inhibition. The effect of MEK inhibition on pancreatic xenografts could be enhanced further by combined application of a PI3K inhibitor. These data provide further rationale for testing combinations of MEK and PI3K inhibitors in clinical trials comprising a patient population with pancreatic cancer harboring mutations in K-RAS.


Sujet(s)
Mitogen-Activated Protein Kinase Kinases/antagonistes et inhibiteurs , Mutation/génétique , Tumeurs du pancréas/enzymologie , Tumeurs du pancréas/génétique , Inhibiteurs des phosphoinositide-3 kinases , Protéines proto-oncogènes/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Protéines G ras/génétique , Animaux , Benzimidazoles/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Extracellular Signal-Regulated MAP Kinases/métabolisme , Femelle , Techniques de knock-down de gènes , Cellules HEK293 , Humains , Indazoles/pharmacologie , Souris , Souris nude , Mitogen-Activated Protein Kinase Kinases/métabolisme , Modèles biologiques , Tumeurs du pancréas/anatomopathologie , Phosphatidylinositol 3-kinases/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes p21(ras) , Sulfonamides/pharmacologie
13.
Mol Cancer Ther ; 11(8): 1747-57, 2012 Aug.
Article de Anglais | MEDLINE | ID: mdl-22653967

RÉSUMÉ

The pan-phosphoinositide 3-kinase (PI3K) inhibitor BKM120 was found, at high concentrations, to cause cell death in various cellular systems, irrespective of their level of PI3K addiction. Transcriptional and biochemical profiling studies were used to identify the origin of these unexpected and apparently PI3K-independent effects. At 5- to 10-fold, the concentration needed to half-maximally inhibit PI3K signaling. BKM120 treatment caused changes in expression of mitotic genes and the induction of a robust G(2)-M arrest. Tubulin polymerization assays and nuclear magnetic resonance-binding studies revealed that BKM120 inhibited microtubule dynamics upon direct binding to tubulin. To assess the contribution of this off-target activity vis-à-vis the antitumor activity of BKM120 in PI3K-dependent tumors, we used a mechanistic PI3K-α-dependent model. We observed that, in vivo, daily treatment of mice with doses of BKM120 up to 40 mg/kg led to tumor regressions with no increase in the mitotic index. Thus, strong antitumor activity can be achieved in PI3K-dependent models at exposures that are below those necessary to engage the off-target activity. In comparison, the clinical data indicate that it is unlikely that BKM120 will achieve exposures sufficient to significantly engage the off-target activity at tolerated doses and schedules. However, in preclinical settings, the consequences of the off-target activity start to manifest themselves at concentrations above 1 µmol/L in vitro and doses above 50 mg/kg in efficacy studies using subcutaneous tumor-bearing mice. Hence, careful concentration and dose range selection is required to ensure that any observation can be correctly attributed to BKM120 inhibition of PI3K.


Sujet(s)
Aminopyridines/pharmacologie , Morpholines/pharmacologie , Inhibiteurs des phosphoinositide-3 kinases , Animaux , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Indazoles/pharmacologie , Souris , Mitose/effets des médicaments et des substances chimiques , Multimérisation de protéines/effets des médicaments et des substances chimiques , Rats , Sulfonamides/pharmacologie , Tubuline/métabolisme
14.
Mol Cancer Ther ; 11(2): 317-28, 2012 Feb.
Article de Anglais | MEDLINE | ID: mdl-22188813

RÉSUMÉ

Following the discovery of NVP-BEZ235, our first dual pan-PI3K/mTOR clinical compound, we sought to identify additional phosphoinositide 3-kinase (PI3K) inhibitors from different chemical classes with a different selectivity profile. The key to achieve these objectives was to couple a structure-based design approach with intensive pharmacologic evaluation of selected compounds during the medicinal chemistry optimization process. Here, we report on the biologic characterization of the 2-morpholino pyrimidine derivative pan-PI3K inhibitor NVP-BKM120. This compound inhibits all four class I PI3K isoforms in biochemical assays with at least 50-fold selectivity against other protein kinases. The compound is also active against the most common somatic PI3Kα mutations but does not significantly inhibit the related class III (Vps34) and class IV (mTOR, DNA-PK) PI3K kinases. Consistent with its mechanism of action, NVP-BKM120 decreases the cellular levels of p-Akt in mechanistic models and relevant tumor cell lines, as well as downstream effectors in a concentration-dependent and pathway-specific manner. Tested in a panel of 353 cell lines, NVP-BKM120 exhibited preferential inhibition of tumor cells bearing PIK3CA mutations, in contrast to either KRAS or PTEN mutant models. NVP-BKM120 shows dose-dependent in vivo pharmacodynamic activity as measured by significant inhibition of p-Akt and tumor growth inhibition in mechanistic xenograft models. NVP-BKM120 behaves synergistically when combined with either targeted agents such as MEK or HER2 inhibitors or with cytotoxic agents such as docetaxel or temozolomide. The pharmacological, biologic, and preclinical safety profile of NVP-BKM120 supports its clinical development and the compound is undergoing phase II clinical trials in patients with cancer.


Sujet(s)
Aminopyridines/pharmacologie , Morpholines/pharmacologie , Tumeurs/traitement médicamenteux , Inhibiteurs des phosphoinositide-3 kinases , Inhibiteurs de protéines kinases/pharmacologie , Administration par voie orale , Aminopyridines/composition chimique , Aminopyridines/pharmacocinétique , Animaux , Biodisponibilité , Technique de Western , Lignée cellulaire tumorale , Relation dose-effet des médicaments , Cellules HCT116 , Cellules HT29 , Humains , Isoenzymes/antagonistes et inhibiteurs , Isoenzymes/génétique , Isoenzymes/métabolisme , Souris , Souris nude , Modèles moléculaires , Structure moléculaire , Morpholines/composition chimique , Morpholines/pharmacocinétique , Mutation , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Phosphatidylinositol 3-kinase/composition chimique , Phosphatidylinositol 3-kinase/métabolisme , Liaison aux protéines , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacocinétique , Structure tertiaire des protéines , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Rats , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE