Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 31
Filtrer
1.
J Clin Oncol ; 41(14): 2651-2660, 2023 05 10.
Article de Anglais | MEDLINE | ID: mdl-36947734

RÉSUMÉ

PURPOSE: No approved targeted therapy for the treatment of patients with neuroblastoma RAS viral (v-ras) oncogene homolog (NRAS)-mutant melanoma is currently available. PATIENTS AND METHODS: In this phase Ib escalation/expansion study (ClinicalTrials.gov identifier: NCT02974725), the safety, tolerability, and preliminary antitumor activity of naporafenib (LXH254), a BRAF/CRAF protein kinases inhibitor, were explored in combination with trametinib in patients with advanced/metastatic KRAS- or BRAF-mutant non-small-cell lung cancer (escalation arm) or NRAS-mutant melanoma (escalation and expansion arms). RESULTS: Thirty-six and 30 patients were enrolled in escalation and expansion, respectively. During escalation, six patients reported grade ≥3 dose-limiting toxicities, including dermatitis acneiform (n = 2), maculopapular rash (n = 2), increased lipase (n = 1), and Stevens-Johnson syndrome (n = 1). The recommended doses for expansion were naporafenib 200 mg twice a day plus trametinib 1 mg once daily and naporafenib 400 mg twice a day plus trametinib 0.5 mg once daily. During expansion, all 30 patients experienced a treatment-related adverse event, the most common being rash (80%, n = 24), blood creatine phosphokinase increased, diarrhea, and nausea (30%, n = 9 each). In expansion, the objective response rate, median duration of response, and median progression-free survival were 46.7% (95% CI, 21.3 to 73.4; 7 of 15 patients), 3.75 (95% CI, 1.97 to not estimable [NE]) months, and 5.52 months, respectively, in patients treated with naporafenib 200 mg twice a day plus trametinib 1 mg once daily, and 13.3% (95% CI, 1.7 to 40.5; 2 of 15 patients), 3.75 (95% CI, 2.04 to NE) months, and 4.21 months, respectively, in patients treated with naporafenib 400 mg twice a day plus trametinib 0.5 mg once daily. CONCLUSION: Naporafenib plus trametinib showed promising preliminary antitumor activity in patients with NRAS-mutant melanoma. Prophylactic strategies aimed to lower the incidence of skin-related events are under investigation.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Exanthème , Tumeurs du poumon , Mélanome , Humains , Protéines proto-oncogènes B-raf/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/traitement médicamenteux , Mélanome/traitement médicamenteux , Mélanome/génétique , Pyridones , Pyrimidinones , Exanthème/induit chimiquement , Exanthème/traitement médicamenteux , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Mutation , Protéines membranaires/génétique , dGTPases/génétique
2.
Cell Rep ; 42(4): 112297, 2023 04 25.
Article de Anglais | MEDLINE | ID: mdl-36961816

RÉSUMÉ

Anti-tumor efficacy of targeted therapies is variable across patients and cancer types. Even in patients with initial deep response, tumors are typically not eradicated and eventually relapse. To address these challenges, we present a systematic screen for targets that limit the anti-tumor efficacy of EGFR and ALK inhibitors in non-small cell lung cancer and BRAF/MEK inhibitors in colorectal cancer. Our approach includes genome-wide CRISPR screens with or without drugs targeting the oncogenic driver ("anchor therapy"), and large-scale pairwise combination screens of anchor therapies with 351 other drugs. Interestingly, targeting of a small number of genes, including MCL1, BCL2L1, and YAP1, sensitizes multiple cell lines to the respective anchor therapy. Data from drug combination screens with EGF816 and ceritinib indicate that dasatinib and agents disrupting microtubules act synergistically across many cell lines. Finally, we show that a higher-order-combination screen with 26 selected drugs in two resistant EGFR-mutant lung cancer cell lines identified active triplet combinations.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Protéines proto-oncogènes B-raf/génétique , Clustered regularly interspaced short palindromic repeats/génétique , Récidive tumorale locale/génétique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Récepteurs ErbB/génétique , Récepteurs à activité tyrosine kinase/génétique , Mitogen-Activated Protein Kinase Kinases/génétique , Mutation , Lignée cellulaire tumorale
3.
Cell Rep ; 40(4): 111095, 2022 07 26.
Article de Anglais | MEDLINE | ID: mdl-35905710

RÉSUMÉ

Reoccurring/high-risk neuroblastoma (NB) tumors have the enrichment of non-RAS/RAF mutations along the mitogen-activated protein kinase (MAPK) signaling pathway, suggesting that activation of MEK/ERK is critical for their survival. However, based on preclinical data, MEK inhibitors are unlikely to be active in NB and have demonstrated dose-limiting toxicities that limit their use. Here, we explore an alternative way to target the MAPK pathway in high-risk NB. We find that NB models are among the most sensitive among over 900 tumor-derived cell lines to the allosteric SHP2 inhibitor SHP099. Sensitivity to SHP099 in NB is greater in models with loss or low expression of the RAS GTPase activation protein (GAP) neurofibromin 1 (NF1). Furthermore, NF1 is lower in advanced and relapsed NB and NF1 loss is enriched in high-risk NB tumors regardless of MYCN status. SHP2 inhibition consistently blocks tumor growth in high-risk NB mouse models, revealing a new drug target in relapsed NB.


Sujet(s)
Neuroblastome , Neurofibromine-1 , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Animaux , Lignée cellulaire tumorale , Souris , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Récidive tumorale locale , Neuroblastome/traitement médicamenteux , Neuroblastome/génétique , Neuroblastome/anatomopathologie , Neurofibromine-1/génétique , Neurofibromine-1/métabolisme , Inhibiteurs de protéines kinases/pharmacologie
4.
Clin Cancer Res ; 27(7): 2061-2073, 2021 04 01.
Article de Anglais | MEDLINE | ID: mdl-33355204

RÉSUMÉ

PURPOSE: Targeting RAF for antitumor therapy in RAS-mutant tumors holds promise. Herein, we describe in detail novel properties of the type II RAF inhibitor, LXH254. EXPERIMENTAL DESIGN: LXH254 was profiled in biochemical, in vitro, and in vivo assays, including examining the activities of the drug in a large panel of cancer-derived cell lines and a comprehensive set of in vivo models. In addition, activity of LXH254 was assessed in cells where different sets of RAF paralogs were ablated, or that expressed kinase-impaired and dimer-deficient variants of ARAF. RESULTS: We describe an unexpected paralog selectivity of LXH254, which is able to potently inhibit BRAF and CRAF, but has less activity against ARAF. LXH254 was active in models harboring BRAF alterations, including atypical BRAF alterations coexpressed with mutant K/NRAS, and NRAS mutants, but had only modest activity in KRAS mutants. In RAS-mutant lines, loss of ARAF, but not BRAF or CRAF, sensitized cells to LXH254. ARAF-mediated resistance to LXH254 required both kinase function and dimerization. Higher concentrations of LXH254 were required to inhibit signaling in RAS-mutant cells expressing only ARAF relative to BRAF or CRAF. Moreover, specifically in cells expressing only ARAF, LXH254 caused paradoxical activation of MAPK signaling in a manner similar to dabrafenib. Finally, in vivo, LXH254 drove complete regressions of isogenic variants of RAS-mutant cells lacking ARAF expression, while parental lines were only modestly sensitive. CONCLUSIONS: LXH254 is a novel RAF inhibitor, which is able to inhibit dimerized BRAF and CRAF, as well as monomeric BRAF, while largely sparing ARAF.


Sujet(s)
Système de signalisation des MAP kinases/physiologie , Tumeurs/traitement médicamenteux , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes B-raf/antagonistes et inhibiteurs , Protéines proto-oncogènes c-raf/antagonistes et inhibiteurs , Animaux , Extracellular Signal-Regulated MAP Kinases/métabolisme , Femelle , Cellules HCT116 , Humains , Souris , Mutation , Tumeurs/génétique , Inhibiteurs de protéines kinases/pharmacologie , Multimérisation de protéines , Protéines proto-oncogènes B-raf/composition chimique , Protéines proto-oncogènes c-raf/composition chimique , Protéines proto-oncogènes p21(ras)/génétique
5.
Clin Cancer Res ; 27(1): 342-354, 2021 01 01.
Article de Anglais | MEDLINE | ID: mdl-33046519

RÉSUMÉ

PURPOSE: SHP2 inhibitors offer an appealing and novel approach to inhibit receptor tyrosine kinase (RTK) signaling, which is the oncogenic driver in many tumors or is frequently feedback activated in response to targeted therapies including RTK inhibitors and MAPK inhibitors. We seek to evaluate the efficacy and synergistic mechanisms of combinations with a novel SHP2 inhibitor, TNO155, to inform their clinical development. EXPERIMENTAL DESIGN: The combinations of TNO155 with EGFR inhibitors (EGFRi), BRAFi, KRASG12Ci, CDK4/6i, and anti-programmed cell death-1 (PD-1) antibody were tested in appropriate cancer models in vitro and in vivo, and their effects on downstream signaling were examined. RESULTS: In EGFR-mutant lung cancer models, combination benefit of TNO155 and the EGFRi nazartinib was observed, coincident with sustained ERK inhibition. In BRAFV600E colorectal cancer models, TNO155 synergized with BRAF plus MEK inhibitors by blocking ERK feedback activation by different RTKs. In KRASG12C cancer cells, TNO155 effectively blocked the feedback activation of wild-type KRAS or other RAS isoforms induced by KRASG12Ci and greatly enhanced efficacy. In addition, TNO155 and the CDK4/6 inhibitor ribociclib showed combination benefit in a large panel of lung and colorectal cancer patient-derived xenografts, including those with KRAS mutations. Finally, TNO155 effectively inhibited RAS activation by colony-stimulating factor 1 receptor, which is critical for the maturation of immunosuppressive tumor-associated macrophages, and showed combination activity with anti-PD-1 antibody. CONCLUSIONS: Our findings suggest TNO155 is an effective agent for blocking both tumor-promoting and immune-suppressive RTK signaling in RTK- and MAPK-driven cancers and their tumor microenvironment. Our data provide the rationale for evaluating these combinations clinically.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonistes et inhibiteurs , Régulation allostérique/effets des médicaments et des substances chimiques , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lignée cellulaire tumorale , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Synergie des médicaments , Récepteurs ErbB/antagonistes et inhibiteurs , Femelle , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Souris , Mutation , Tumeurs/génétique , Tumeurs/immunologie , Tumeurs/anatomopathologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes B-raf/antagonistes et inhibiteurs , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes p21(ras)/antagonistes et inhibiteurs , Protéines proto-oncogènes p21(ras)/génétique , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe
7.
Oncotarget ; 11(3): 265-281, 2020 Jan 21.
Article de Anglais | MEDLINE | ID: mdl-32076487

RÉSUMÉ

SHP2 mediates RAS activation downstream of multiple receptor tyrosine kinases (RTKs) and cancer cell lines dependent on RTKs are in general dependent on SHP2. Profiling of the allosteric SHP2 inhibitor SHP099 across cancer cell lines harboring various RTK dependencies reveals that FGFR-dependent cells are often insensitive to SHP099 when compared to EGFR-dependent cells. We find that FGFR-driven cells depend on SHP2 but exhibit resistance to SHP2 inhibitors in vitro and in vivo. Treatment of such models with SHP2 inhibitors results in an initial decrease in phosphorylated ERK1/2 (p-ERK) levels, however p-ERK levels rapidly rebound within two hours. This p-ERK rebound is blocked by FGFR inhibitors or high doses of SHP2 inhibitors. Mechanistically, compared with EGFR-driven cells, FGFR-driven cells tend to express high levels of RTK negative regulators such as the SPRY family proteins, which are rapidly downregulated upon ERK inhibition. Moreover, over-expression of SPRY4 in FGFR-driven cells prevents MAPK pathway reactivation and sensitizes them to SHP2 inhibitors. We also identified two novel combination approaches to enhance the efficacy of SHP2 inhibitors, either with a distinct site 2 allosteric SHP2 inhibitor or with a RAS-SOS1 interaction inhibitor. Our findings suggest the rapid FGFR feedback activation following initial pathway inhibition by SHP2 inhibitors may promote the open conformation of SHP2 and lead to resistance to SHP2 inhibitors. These findings may assist to refine patient selection and predict resistance mechanisms in the clinical development of SHP2 inhibitors and to suggest strategies for discovering SHP2 inhibitors that are more effective against upstream feedback activation.

8.
Mol Cancer Ther ; 18(12): 2368-2380, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31439712

RÉSUMÉ

KRAS, an oncogene mutated in nearly one third of human cancers, remains a pharmacologic challenge for direct inhibition except for recent advances in selective inhibitors targeting the G12C variant. Here, we report that selective inhibition of the protein tyrosine phosphatase, SHP2, can impair the proliferation of KRAS-mutant cancer cells in vitro and in vivo using cell line xenografts and primary human tumors. In vitro, sensitivity of KRAS-mutant cells toward the allosteric SHP2 inhibitor, SHP099, is not apparent when cells are grown on plastic in 2D monolayer, but is revealed when cells are grown as 3D multicellular spheroids. This antitumor activity is also observed in vivo in mouse models. Interrogation of the MAPK pathway in SHP099-treated KRAS-mutant cancer models demonstrated similar modulation of p-ERK and DUSP6 transcripts in 2D, 3D, and in vivo, suggesting a MAPK pathway-dependent mechanism and possible non-MAPK pathway-dependent mechanisms in tumor cells or tumor microenvironment for the in vivo efficacy. For the KRASG12C MIAPaCa-2 model, we demonstrate that the efficacy is cancer cell intrinsic as there is minimal antiangiogenic activity by SHP099, and the effects of SHP099 is recapitulated by genetic depletion of SHP2 in cancer cells. Furthermore, we demonstrate that SHP099 efficacy in KRAS-mutant models can be recapitulated with RTK inhibitors, suggesting RTK activity is responsible for the SHP2 activation. Taken together, these data reveal that many KRAS-mutant cancers depend on upstream signaling from RTK and SHP2, and provide a new therapeutic framework for treating KRAS-mutant cancers with SHP2 inhibitors.


Sujet(s)
Tumeurs/traitement médicamenteux , Tumeurs/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonistes et inhibiteurs , Protéines proto-oncogènes p21(ras)/génétique , Tachykinines/antagonistes et inhibiteurs , Animaux , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Femelle , Humains , Souris , Tumeurs/anatomopathologie , Transduction du signal , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Mol Cancer Ther ; 18(7): 1323-1334, 2019 07.
Article de Anglais | MEDLINE | ID: mdl-31068384

RÉSUMÉ

FGFR1 was recently shown to be activated as part of a compensatory response to prolonged treatment with the MEK inhibitor trametinib in several KRAS-mutant lung and pancreatic cancer cell lines. We hypothesize that other receptor tyrosine kinases (RTK) are also feedback-activated in this context. Herein, we profile a large panel of KRAS-mutant cancer cell lines for the contribution of RTKs to the feedback activation of phospho-MEK following MEK inhibition, using an SHP2 inhibitor (SHP099) that blocks RAS activation mediated by multiple RTKs. We find that RTK-driven feedback activation widely exists in KRAS-mutant cancer cells, to a less extent in those harboring the G13D variant, and involves several RTKs, including EGFR, FGFR, and MET. We further demonstrate that this pathway feedback activation is mediated through mutant KRAS, at least for the G12C, G12D, and G12V variants, and wild-type KRAS can also contribute significantly to the feedback activation. Finally, SHP099 and MEK inhibitors exhibit combination benefits inhibiting KRAS-mutant cancer cell proliferation in vitro and in vivo These findings provide a rationale for exploration of combining SHP2 and MAPK pathway inhibitors for treating KRAS-mutant cancers in the clinic.


Sujet(s)
Acrylonitrile/analogues et dérivés , Dérivés de l'aniline/usage thérapeutique , Tumeurs/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Protéines proto-oncogènes p21(ras)/génétique , Acrylonitrile/pharmacologie , Acrylonitrile/usage thérapeutique , Dérivés de l'aniline/pharmacologie , Animaux , Lignée cellulaire tumorale , Femelle , Humains , Souris , Souris nude , Tumeurs/métabolisme , Transfection , Tests d'activité antitumorale sur modèle de xénogreffe
10.
Nature ; 569(7757): 503-508, 2019 05.
Article de Anglais | MEDLINE | ID: mdl-31068700

RÉSUMÉ

Large panels of comprehensively characterized human cancer models, including the Cancer Cell Line Encyclopedia (CCLE), have provided a rigorous framework with which to study genetic variants, candidate targets, and small-molecule and biological therapeutics and to identify new marker-driven cancer dependencies. To improve our understanding of the molecular features that contribute to cancer phenotypes, including drug responses, here we have expanded the characterizations of cancer cell lines to include genetic, RNA splicing, DNA methylation, histone H3 modification, microRNA expression and reverse-phase protein array data for 1,072 cell lines from individuals of various lineages and ethnicities. Integration of these data with functional characterizations such as drug-sensitivity, short hairpin RNA knockdown and CRISPR-Cas9 knockout data reveals potential targets for cancer drugs and associated biomarkers. Together, this dataset and an accompanying public data portal provide a resource for the acceleration of cancer research using model cancer cell lines.


Sujet(s)
Lignée cellulaire tumorale , Tumeurs/génétique , Tumeurs/anatomopathologie , Antinéoplasiques/pharmacologie , Marqueurs biologiques tumoraux , Méthylation de l'ADN , Résistance aux médicaments antinéoplasiques , Ethnies/génétique , Édition de gène , Histone/métabolisme , Humains , microARN/génétique , Thérapie moléculaire ciblée , Tumeurs/métabolisme , Analyse par réseau de protéines , Épissage des ARN
12.
Oncotarget ; 9(81): 35226-35240, 2018 Oct 16.
Article de Anglais | MEDLINE | ID: mdl-30443290

RÉSUMÉ

Inhibition of cyclin-dependent kinases 4 and 6 (CDK4/6) is associated with robust antitumor activity. Ribociclib (LEE011) is an orally bioavailable CDK4/6 inhibitor that is approved for the treatment of hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer, in combination with an aromatase inhibitor, and is currently being evaluated in several additional trials. Here, we report the preclinical profile of ribociclib. When tested across a large panel of kinase active site binding assays, ribociclib and palbociclib were highly selective for CDK4, while abemaciclib showed affinity to several other kinases. Both ribociclib and abemaciclib showed slightly higher potency in CDK4-dependent cells than in CDK6-dependent cells, while palbociclib did not show such a difference. Profiling CDK4/6 inhibitors in large-scale cancer cell line screens in vitro confirmed that RB1 loss of function is a negative predictor of sensitivity. We also found that routinely used cellular viability assays measuring adenosine triphosphate levels as a proxy for cell numbers underestimated the effects of CDK4/6 inhibition, which contrasts with assays that assess cell number more directly. Robust antitumor efficacy and combination benefit was detected when ribociclib was added to encorafenib, nazartinib, or endocrine therapies in patient-derived xenografts.

13.
Nat Med ; 24(4): 512-517, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-29505033

RÉSUMÉ

Most anaplastic lymphoma kinase (ALK)-rearranged non-small-cell lung tumors initially respond to small-molecule ALK inhibitors, but drug resistance often develops. Of tumors that develop resistance to highly potent second-generation ALK inhibitors, approximately half harbor resistance mutations in ALK, while the other half have other mechanisms underlying resistance. Members of the latter group often have activation of at least one of several different tyrosine kinases driving resistance. Such tumors are not expected to respond to lorlatinib-a third-generation inhibitor targeting ALK that is able to overcome all clinically identified resistant mutations in ALK-and further therapeutic options are limited. Herein, we deployed a shRNA screen of 1,000 genes in multiple ALK-inhibitor-resistant patient-derived cells (PDCs) to discover those that confer sensitivity to ALK inhibition. This approach identified SHP2, a nonreceptor protein tyrosine phosphatase, as a common targetable resistance node in multiple PDCs. SHP2 provides a parallel survival input downstream of multiple tyrosine kinases that promote resistance to ALK inhibitors. Treatment with SHP099, the recently discovered small-molecule inhibitor of SHP2, in combination with the ALK tyrosine kinase inhibitor (TKI) ceritinib halted the growth of resistant PDCs through preventing compensatory RAS and ERK1 and ERK2 (ERK1/2) reactivation. These findings suggest that combined ALK and SHP2 inhibition may be a promising therapeutic strategy for resistant cancers driven by several different ALK-independent mechanisms underlying resistance.


Sujet(s)
Kinase du lymphome anaplasique/antagonistes et inhibiteurs , Carcinome pulmonaire non à petites cellules/enzymologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Réarrangement des gènes/génétique , Tumeurs du poumon/enzymologie , Inhibiteurs de protéines kinases/pharmacologie , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonistes et inhibiteurs , Kinase du lymphome anaplasique/métabolisme , Animaux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Extracellular Signal-Regulated MAP Kinases/métabolisme , Femelle , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Souris nude , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Pyrimidines/pharmacologie , Pyrimidines/usage thérapeutique , Petit ARN interférent/métabolisme , Sulfones/pharmacologie , Sulfones/usage thérapeutique
14.
Cancer Discov ; 8(5): 648-661, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-29483135

RÉSUMÉ

Mutations at multiple sites in MEK1 occur in cancer, suggesting that their mechanisms of activation might be different. We analyzed 17 tumor-associated MEK1 mutants and found that they drove ERK signaling autonomously or in a RAS/RAF-dependent manner. The latter are sensitive to feedback inhibition of RAF, which limits their functional output, and often cooccur with RAS or RAF mutations. They act as amplifiers of RAF signaling. In contrast, another class of mutants deletes a hitherto unrecognized negative regulatory segment of MEK1, is RAF- and phosphorylation-independent, is unaffected by feedback inhibition of upstream signaling, and drives high ERK output and transformation in the absence of RAF activity. Moreover, these RAF-independent mutants are insensitive to allosteric MEK inhibitors, which preferentially bind to the inactivated form of MEK1. All the mutants are sensitive to an ATP-competitive MEK inhibitor. Thus, our study comprises a novel therapeutic strategy for tumors driven by RAF-independent MEK1 mutants.Significance: Mutants with which MEK1 mutants coexist and their sensitivity to inhibitors are determined by allele-specific properties. This study shows the importance of functional characterization of mutant alleles in single oncogenes and identifies a new class of MEK1 mutants, insensitive to current MEK1 inhibitors but treatable with a new ATP-competitive inhibitor. Cancer Discov; 8(5); 648-61. ©2018 AACR.See related commentary by Maust et al., p. 534This article is highlighted in the In This Issue feature, p. 517.


Sujet(s)
Allèles , MAP Kinase Kinase 1/génétique , Mutation , Adénosine triphosphate/métabolisme , Animaux , Lignée cellulaire , Résistance aux médicaments antinéoplasiques/génétique , Humains , MAP Kinase Kinase 1/composition chimique , Souris , Tumeurs/génétique , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Phosphorylation , Liaison aux protéines , Conformation des protéines , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes c-raf/métabolisme , Délétion de séquence , Transduction du signal/effets des médicaments et des substances chimiques , Kinases raf/métabolisme
15.
Cancer Res ; 78(6): 1537-1548, 2018 03 15.
Article de Anglais | MEDLINE | ID: mdl-29343524

RÉSUMÉ

Resistance to the RAF inhibitor vemurafenib arises commonly in melanomas driven by the activated BRAF oncogene. Here, we report antitumor properties of RAF709, a novel ATP-competitive kinase inhibitor with high potency and selectivity against RAF kinases. RAF709 exhibited a mode of RAF inhibition distinct from RAF monomer inhibitors such as vemurafenib, showing equal activity against both RAF monomers and dimers. As a result, RAF709 inhibited MAPK signaling activity in tumor models harboring either BRAFV600 alterations or mutant N- and KRAS-driven signaling, with minimal paradoxical activation of wild-type RAF. In cell lines and murine xenograft models, RAF709 demonstrated selective antitumor activity in tumor cells harboring BRAF or RAS mutations compared with cells with wild-type BRAF and RAS genes. RAF709 demonstrated a direct pharmacokinetic/pharmacodynamic relationship in in vivo tumor models harboring KRAS mutation. Furthermore, RAF709 elicited regression of primary human tumor-derived xenograft models with BRAF, NRAS, or KRAS mutations with excellent tolerability. Our results support further development of inhibitors like RAF709, which represents a next-generation RAF inhibitor with unique biochemical and cellular properties that enables antitumor activities in RAS-mutant tumors.Significance: In an effort to develop RAF inhibitors with the appropriate pharmacological properties to treat RAS mutant tumors, RAF709, a compound with potency, selectivity, and in vivo properties, was developed that will allow preclinical therapeutic hypothesis testing, but also provide an excellent probe to further unravel the complexities of RAF kinase signaling. Cancer Res; 78(6); 1537-48. ©2018 AACR.


Sujet(s)
2,2'-Bipyridine/analogues et dérivés , Antinéoplasiques/pharmacologie , Benzamides/pharmacologie , Protéines proto-oncogènes B-raf/génétique , Kinases raf/antagonistes et inhibiteurs , Protéines G ras/génétique , 2,2'-Bipyridine/pharmacologie , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Femelle , Humains , MAP Kinase Kinase Kinases/antagonistes et inhibiteurs , Souris nude , Mutation , Inhibiteurs de protéines kinases/pharmacologie , Multimérisation de protéines , Tests d'activité antitumorale sur modèle de xénogreffe , Kinases raf/métabolisme
16.
Cell Rep ; 21(7): 1953-1967, 2017 Nov 14.
Article de Anglais | MEDLINE | ID: mdl-29141225

RÉSUMÉ

Therapy of advanced melanoma is changing dramatically. Following mutational and biological subclassification of this heterogeneous cancer, several targeted and immune therapies were approved and increased survival significantly. To facilitate further advancements through pre-clinical in vivo modeling, we have established 459 patient-derived xenografts (PDX) and live tissue samples from 384 patients representing the full spectrum of clinical, therapeutic, mutational, and biological heterogeneity of melanoma. PDX have been characterized using targeted sequencing and protein arrays and are clinically annotated. This exhaustive live tissue resource includes PDX from 57 samples resistant to targeted therapy, 61 samples from responders and non-responders to immune checkpoint blockade, and 31 samples from brain metastasis. Uveal, mucosal, and acral subtypes are represented as well. We show examples of pre-clinical trials that highlight how the PDX collection can be used to develop and optimize precision therapies, biomarkers of response, and the targeting of rare genetic subgroups.


Sujet(s)
Hétérogreffes/anatomopathologie , Mélanome/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes , Animaux , Cellules cultivées , Hétérogreffes/métabolisme , Humains , Mélanome/classification , Mélanome/génétique , Souris
17.
Clin Cancer Res ; 23(18): 5339-5348, 2017 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-28611198

RÉSUMÉ

Purpose: Encorafenib, a selective BRAF inhibitor (BRAFi), has a pharmacologic profile that is distinct from that of other clinically active BRAFis. We evaluated encorafenib in a phase I study in patients with BRAFi treatment-naïve and pretreated BRAF-mutant melanoma.Experimental Design: The pharmacologic activity of encorafenib was first characterized preclinically. Encorafenib monotherapy was then tested across a range of once-daily (50-700 mg) or twice-daily (75-150 mg) regimens in a phase I, open-label, dose-escalation and -expansion study in adult patients with histologically confirmed advanced/metastatic BRAF-mutant melanoma. Study objectives were to determine the maximum tolerated dose (MTD) and/or recommended phase II dose (RP2D), characterize the safety and tolerability and pharmacokinetic profile, and assess the preliminary antitumor activity of encorafenib.Results: Preclinical data demonstrated that encorafenib inhibited BRAF V600E kinase activity with a prolonged off-rate and suppressed proliferation and tumor growth of BRAF V600E-mutant melanoma models. In the dose-escalation phase, 54 patients (29 BRAFi-pretreated and 25 BRAFi-naïve) were enrolled. Seven patients in the dose-determining set experienced dose-limiting toxicities. Encorafenib at a dose of 300 mg once daily was declared the RP2D. In the expansion phase, the most common all-cause adverse events were nausea (66%), myalgia (63%), and palmar-plantar erythrodysesthesia (54%). In BRAFi-naïve patients, the overall response rate (ORR) and median progression-free survival (mPFS) were 60% and 12.4 months [95% confidence interval (CI), 7.4-not reached (NR)]. In BRAFi-pretreated patients, the ORR and mPFS were 22% and 1.9 months (95% CI, 0.9-3.7).Conclusions: Once-daily dosing of single-agent encorafenib had a distinct tolerability profile and showed varying antitumor activity across BRAFi-pretreated and BRAFi-naïve patients with advanced/metastatic melanoma. Clin Cancer Res; 23(18); 5339-48. ©2017 AACR.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Carbamates/usage thérapeutique , Mélanome/traitement médicamenteux , Mélanome/génétique , Mutation , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes B-raf/antagonistes et inhibiteurs , Protéines proto-oncogènes B-raf/génétique , Sulfonamides/usage thérapeutique , Animaux , Antinéoplasiques/administration et posologie , Antinéoplasiques/effets indésirables , Antinéoplasiques/pharmacocinétique , Carbamates/administration et posologie , Carbamates/effets indésirables , Carbamates/pharmacocinétique , Modèles animaux de maladie humaine , Évaluation préclinique de médicament , Surveillance des médicaments , Femelle , Humains , Estimation de Kaplan-Meier , Mâle , Dose maximale tolérée , Mélanome/mortalité , Mélanome/anatomopathologie , Souris , Thérapie moléculaire ciblée , Stadification tumorale , Inhibiteurs de protéines kinases/administration et posologie , Inhibiteurs de protéines kinases/effets indésirables , Inhibiteurs de protéines kinases/pharmacocinétique , Sulfonamides/administration et posologie , Sulfonamides/effets indésirables , Sulfonamides/pharmacocinétique , Résultat thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe
18.
Mol Cancer Res ; 15(10): 1431-1444, 2017 10.
Article de Anglais | MEDLINE | ID: mdl-28655712

RÉSUMÉ

Alterations in MEK1/2 occur in cancers, both in the treatment-naïve state and following targeted therapies, most notably BRAF and MEK inhibitors in BRAF-V600E-mutant melanoma and colorectal cancer. Efforts were undertaken to understand the effects of these mutations, based upon protein structural location, and MEK1/2 activity. Two categories of MEK1/2 alterations were evaluated, those associated with either the allosteric pocket or helix-A. Clinically, MEK1/2 alterations of the allosteric pocket are rare and we demonstrate that they confer resistance to MEK inhibitors, while retaining sensitivity to BRAF inhibition. Most mutations described in patients fall within, or are associated with, helix-A. Mutations in this region reduce sensitivity to both BRAF and MEK inhibition and display elevated phospho-ERK1/2 levels, independent from increases in phospho-MEK1/2. Biochemical experiments with a representative helix-A variant, MEK1-Q56P, reveal both increased catalytic efficiency of the activated enzyme, and phosphorylation-independent activity relative to wild-type MEK1. Consistent with these findings, MEK1/2 alterations in helix A retain sensitivity to downstream antagonism via pharmacologic inhibition of ERK1/2. This work highlights the importance of classifying mutations based on structural and phenotypic consequences, both in terms of pathway signaling output and response to pharmacologic inhibition.Implications: This study suggests that alternate modes of target inhibition, such as ERK inhibition, will be required to effectively treat tumors harboring these MEK1/2-resistant alleles. Mol Cancer Res; 15(10); 1431-44. ©2017 AACR.


Sujet(s)
Tumeurs colorectales/génétique , MAP Kinase Kinase 1/génétique , MAP Kinase Kinase 2/génétique , Mutation , Inhibiteurs de protéines kinases/pharmacologie , Kinases raf/métabolisme , Site allostérique , Lignée cellulaire tumorale , Prolifération cellulaire , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , MAP Kinase Kinase 1/composition chimique , MAP Kinase Kinase 1/métabolisme , MAP Kinase Kinase 2/composition chimique , MAP Kinase Kinase 2/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Modèles moléculaires , Phosphorylation , Structure secondaire des protéines , Protéines proto-oncogènes B-raf/composition chimique , Protéines proto-oncogènes B-raf/génétique
19.
Clin Cancer Res ; 23(11): 2856-2868, 2017 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-27986745

RÉSUMÉ

Purpose: Anaplastic lymphoma kinase (ALK) is the most frequently mutated oncogene in the pediatric cancer neuroblastoma. We performed an in vitro screen for synergistic drug combinations that target neuroblastomas with mutations in ALK to determine whether drug combinations could enhance antitumor efficacy.Experimental Design: We screened combinations of eight molecularly targeted agents against 17 comprehensively characterized human neuroblastoma-derived cell lines. We investigated the combination of ceritinib and ribociclib on in vitro proliferation, cell cycle, viability, caspase activation, and the cyclin D/CDK4/CDK6/RB and pALK signaling networks in cell lines with representative ALK status. We performed in vivo trials in CB17 SCID mice bearing conventional and patient-derived xenograft models comparing ceritinib alone, ribociclib alone, and the combination, with plasma pharmacokinetics to evaluate for drug-drug interactions.Results: The combination of ribociclib, a dual inhibitor of cyclin-dependent kinase (CDK) 4 and 6, and the ALK inhibitor ceritinib demonstrated higher cytotoxicity (P = 0.008) and synergy scores (P = 0.006) in cell lines with ALK mutations as compared with cell lines lacking mutations or alterations in ALK Compared with either drug alone, combination therapy enhanced growth inhibition, cell-cycle arrest, and caspase-independent cell death. Combination therapy achieved complete regressions in neuroblastoma xenografts with ALK-F1174L and F1245C de novo resistance mutations and prevented the emergence of resistance. Murine ribociclib and ceritinib plasma concentrations were unaltered by combination therapy.Conclusions: This preclinical combination drug screen with in vivo validation has provided the rationale for a first-in-children trial of combination ceritinib and ribociclib in a molecularly selected pediatric population. Clin Cancer Res; 23(11); 2856-68. ©2016 AACR.


Sujet(s)
Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Neuroblastome/traitement médicamenteux , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Aminopyridines/administration et posologie , Kinase du lymphome anaplasique , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cycline D/génétique , Kinase-4 cycline-dépendante/génétique , Kinase-6 cycline-dépendante/génétique , Synergie des médicaments , Humains , Souris , Mutation , Neuroblastome/génétique , Neuroblastome/anatomopathologie , Purines/administration et posologie , Pyrimidines/administration et posologie , Récepteurs à activité tyrosine kinase/génétique , Protéine du rétinoblastome/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Bibliothèques de petites molécules/administration et posologie , Sulfones/administration et posologie , Tests d'activité antitumorale sur modèle de xénogreffe
20.
Clin Cancer Res ; 23(7): 1785-1796, 2017 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-27729458

RÉSUMÉ

Purpose: Neuroblastoma is treated with aggressive multimodal therapy, yet more than 50% of patients experience relapse. We recently showed that relapsed neuroblastomas frequently harbor mutations leading to hyperactivated ERK signaling and sensitivity to MEK inhibition therapy. Here we sought to define a synergistic therapeutic partner to potentiate MEK inhibition.Experimental Design: We first surveyed 22 genetically annotated human neuroblastoma-derived cell lines (from 20 unique patients) for sensitivity to the MEK inhibitor binimetinib. After noting an inverse correlation with sensitivity to ribociclib (CDK4/6 inhibitor), we studied the combinatorial effect of these two agents using proliferation assays, cell-cycle analysis, Ki67 immunostaining, time-lapse microscopy, and xenograft studies.Results: Sensitivity to binimetinib and ribociclib was inversely related (r = -0.58, P = 0.009). MYCN amplification status and expression were associated with ribociclib sensitivity and binimetinib resistance, whereas increased MAPK signaling was the main determinant of binimetinib sensitivity and ribociclib resistance. Treatment with both compounds resulted in synergistic or additive cellular growth inhibition in all lines tested and significant inhibition of tumor growth in three of four xenograft models of neuroblastoma. The augmented growth inhibition was attributed to diminished cell-cycle progression that was reversible upon removal of drugs.Conclusions: Here we demonstrate that combined binimetinib and ribociclib treatment shows therapeutic synergy across a broad panel of high-risk neuroblastoma preclinical models. These data support testing this combination therapy in relapsed high-risk neuroblastoma patients, with focus on cases with hyperactivated RAS-MAPK signaling. Clin Cancer Res; 23(7); 1785-96. ©2016 AACR.


Sujet(s)
Résistance aux médicaments antinéoplasiques/génétique , Récidive tumorale locale/traitement médicamenteux , Neuroblastome/traitement médicamenteux , Inhibiteurs de protéines kinases/administration et posologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Benzimidazoles/administration et posologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Humains , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Souris , Récidive tumorale locale/génétique , Récidive tumorale locale/anatomopathologie , Neuroblastome/génétique , Neuroblastome/anatomopathologie , Phosphorylation , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...