Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 33
Filtrer
3.
Blood ; 141(6): 634-644, 2023 02 09.
Article de Anglais | MEDLINE | ID: mdl-36219880

RÉSUMÉ

Randomized trials in acute myeloid leukemia (AML) have demonstrated improved survival by the BCL-2 inhibitor venetoclax combined with azacitidine in older patients, and clinical trials are actively exploring the role of venetoclax in combination with intensive chemotherapy in fitter patients with AML. As most patients still develop recurrent disease, improved understanding of relapse mechanisms is needed. We find that 17% of patients relapsing after venetoclax-based therapy for AML have acquired inactivating missense or frameshift/nonsense mutations in the apoptosis effector gene BAX. In contrast, such variants were rare after genotoxic chemotherapy. BAX variants arose within either leukemic or preleukemic compartments, with multiple mutations observed in some patients. In vitro, AML cells with mutated BAX were competitively selected during prolonged exposure to BCL-2 antagonists. In model systems, AML cells rendered deficient for BAX, but not its close relative BAK, displayed resistance to BCL-2 targeting, whereas sensitivity to conventional chemotherapy was variable. Acquired mutations in BAX during venetoclax-based therapy represent a novel mechanism of resistance to BH3-mimetics and a potential barrier to the long-term efficacy of drugs targeting BCL-2 in AML.


Sujet(s)
Leucémie aigüe myéloïde , Protéines proto-oncogènes c-bcl-2 , Humains , Sujet âgé , Protéine Bax/génétique , Lignée cellulaire tumorale , Protéines proto-oncogènes c-bcl-2/génétique , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Composés hétérocycliques bicycliques/usage thérapeutique , Composés hétérocycliques bicycliques/pharmacologie , Apoptose , Mutation
4.
Clin Cancer Res ; 28(6): 1087-1097, 2022 03 15.
Article de Anglais | MEDLINE | ID: mdl-34921024

RÉSUMÉ

PURPOSE: Well-differentiated (WDLPS) and dedifferentiated (DDLPS) liposarcoma are characterized by co-amplification of the murine double minute-2 (MDM2) and cyclin-dependent kinase-4 (CDK4) oncogenes. Siremadlin, a p53-MDM2 inhibitor, was combined with ribociclib, a CDK4/6 inhibitor, in patients with locally advanced/metastatic WDLPS or DDLPS who had radiologically progressed on, or despite, prior systemic therapy. PATIENTS AND METHODS: In this proof-of-concept, phase Ib, dose-escalation study, patients received siremadlin and ribociclib across different regimens until unacceptable toxicity, disease progression, and/or treatment discontinuation: Regimen A [4-week cycle: siremadlin once daily (QD) and ribociclib QD (2 weeks on, 2 weeks off)], Regimen B [3-week cycle: siremadlin once every 3 weeks; ribociclib QD (2 weeks on, 1 week off)], and Regimen C [4-week cycle: siremadlin once every 4 weeks; ribociclib QD (2 weeks on, 2 weeks off)]. The primary objective was to determine the maximum tolerated dose (MTD) and/or recommended dose for expansion (RDE) of siremadlin plus ribociclib in one or more regimens. RESULTS: As of October 16, 2019 (last patient last visit), 74 patients had enrolled. Median duration of exposure was 13 (range, 1-174) weeks. Dose-limiting toxicities occurred in 10 patients, most of which were Grade 3/4 hematologic events. The RDE was siremadlin 120 mg every 3 weeks plus ribociclib 200 mg QD (Regimen B). Three patients achieved a partial response, and 38 achieved stable disease. One patient (Regimen C) died as a result of treatment-related hematotoxicity. CONCLUSIONS: Siremadlin plus ribociclib demonstrated manageable toxicity and early signs of antitumor activity in patients with advanced WDLPS or DDLPS.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Liposarcome , Aminopyridines/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Kinase-4 cycline-dépendante/génétique , Humains , Imidazoles/usage thérapeutique , Liposarcome/traitement médicamenteux , Liposarcome/génétique , Liposarcome/anatomopathologie , Protéines proto-oncogènes c-mdm2/génétique , Purines/usage thérapeutique , Pyrimidines/usage thérapeutique , Pyrroles/usage thérapeutique
5.
Clin Cancer Res ; 28(5): 870-881, 2022 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-34862243

RÉSUMÉ

PURPOSE: This phase I, dose-escalation study investigated the recommended dose for expansion (RDE) of siremadlin, a p53-MDM2 inhibitor, in patients with wild-type TP53 advanced solid or hematologic cancers. PATIENTS AND METHODS: Initial dosing regimens were: 1A (day 1; 21-day cycle; dose 12.5-350 mg) and 2A (days 1-14; 28-day cycle; dose 1-20 mg). Alternative regimens included 1B (days 1 and 8; 28-day cycle) and 2C (days 1-7; 28-day cycle). The primary endpoint was incidence of dose-limiting toxicities (DLT) during cycle 1. RESULTS: Overall, 115 patients with solid tumors and 93 with hematologic malignancies received treatment. DLTs occurred in 8/92 patients with solid tumors and 10/53 patients with hematologic malignancies. In solid tumors, an RDE of 120 mg was defined in 1B. In hematologic tumors, RDEs were defined in 1A: 250 mg, 1B: 120 mg, and 2C: 45 mg. More patients with hematologic malignancies compared with solid tumors experienced grade 3/4 treatment-related adverse events (71% vs. 45%), most commonly resulting from myelosuppression. These were more frequent and severe in patients with hematologic malignancies; 22 patients exhibited tumor lysis syndrome. Overall response rates at the RDEs were 10.3% [95% confidence interval (CI), 2.2-27.4] in solid tumors and 4.2% (95% CI, 0.1-21.1), 20% (95% CI, 4.3-48.1), and 22.2% (95% CI, 8.6-42.3) in acute myeloid leukemia (AML) in 1B, 1A, and 2C, respectively. CONCLUSIONS: A common safety profile was identified and preliminary activity was noted, particularly in AML. Comprehensive investigation of dosing regimens yielded recommended doses/regimens for future combination studies.


Sujet(s)
Tumeurs hématologiques , Leucémie aigüe myéloïde , Tumeurs , Relation dose-effet des médicaments , Tumeurs hématologiques/traitement médicamenteux , Tumeurs hématologiques/génétique , Humains , Imidazoles , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Dose maximale tolérée , Tumeurs/traitement médicamenteux , Pyrimidines , Pyrroles , Protéine p53 suppresseur de tumeur/génétique
6.
Br J Cancer ; 125(5): 687-698, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-34140638

RÉSUMÉ

BACKGROUND: CGM097 inhibits the p53-HDM2 interaction leading to downstream p53 activation. Preclinical in vivo studies support clinical exploration while providing preliminary evidence for dosing regimens. This first-in-human phase I study aimed at assessing the safety, MTD, PK/PD and preliminary antitumor activity of CGM097 in advanced solid tumour patients (NCT01760525). METHODS: Fifty-one patients received oral treatment with CGM097 10-400 mg 3qw (n = 31) or 300-700 mg 3qw 2 weeks on/1 week off (n = 20). Choice of dose regimen was guided by PD biomarkers, and quantitative models describing the effect of CGM097 on circulating platelet and PD kinetics. RESULTS: No dose-limiting toxicities were reported in any regimens. The most common treatment-related grade 3/4 AEs were haematologic events. PK/PD models well described the time course of platelet and serum GDF-15 changes, providing a tool to predict response to CGM097 for dose-limiting thrombocytopenia and GDF-15 biomarker. The disease control rate was 39%, including one partial response and 19 patients in stable disease. Twenty patients had a cumulative treatment duration of >16 weeks, with eight patients on treatment for >32 weeks. The MTD was not determined. CONCLUSIONS: Despite delayed-onset thrombocytopenia frequently observed, the tolerability of CGM097 appears manageable. This study provided insights on dosing optimisation for next-generation HDM2 inhibitors. TRANSLATIONAL RELEVANCE: Haematologic toxicity with delayed thrombocytopenia is a well-known on-target effect of HDM2 inhibitors. Here we have developed a PK/PD guided approach to optimise the dose and schedule of CGM097, a novel HDM2 inhibitor, using exposure, platelets and GDF-15, a known p53 downstream target to predict patients at higher risk to develop thrombocytopenia. While CGM097 had shown limited activity, with disease control rate of 39% and only one patient in partial response, the preliminary data from the first-in-human escalation study together with the PK/PD modeling provide important insights on how to optimize dosing of next generation HDM2 inhibitors to mitigate hematologic toxicity.


Sujet(s)
Facteur-15 de croissance et de différenciation/sang , Isoquinoléines/administration et posologie , Tumeurs/traitement médicamenteux , Pipérazines/administration et posologie , Administration par voie orale , Adulte , Sujet âgé , Animaux , Marqueurs biologiques tumoraux/sang , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire , Calendrier d'administration des médicaments , Calcul des posologies , Femelle , Humains , Isoquinoléines/effets indésirables , Isoquinoléines/pharmacocinétique , Mâle , Souris , Adulte d'âge moyen , Tumeurs/sang , Pipérazines/effets indésirables , Pipérazines/pharmacocinétique , Résultat thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe
7.
Cancer Res ; 81(11): 3079-3091, 2021 06 01.
Article de Anglais | MEDLINE | ID: mdl-33504557

RÉSUMÉ

p53 is a transcription factor that plays a central role in guarding the genomic stability of cells through cell-cycle arrest or induction of apoptosis. However, the effects of p53 in antitumor immunity are poorly understood. To investigate the role of p53 in controlling tumor-immune cell cross-talk, we studied murine syngeneic models treated with HDM201, a potent and selective second-generation MDM2 inhibitor. In response to HDM201 treatment, the percentage of dendritic cells increased, including the CD103+ antigen cross-presenting subset. Furthermore, HDM201 increased the percentage of Tbet+Eomes+ CD8+ T cells and the CD8+/Treg ratio within the tumor. These immunophenotypic changes were eliminated with the knockout of p53 in tumor cells. Enhanced expression of CD80 on tumor cells was observed in vitro and in vivo, which coincided with T-cell-mediated tumor cell killing. Combining HDM201 with PD-1 or PD-L1 blockade increased the number of complete tumor regressions. Responding mice developed durable, antigen-specific memory T cells and rejected subsequent tumor implantation. Importantly, antitumor activity of HDM201 in combination with PD-1/PD-L1 blockade was abrogated in p53-mutated and knockout syngeneic tumor models, indicating the effect of HDM201 on the tumor is required for triggering antitumor immunity. Taken together, these results demonstrate that MDM2 inhibition triggers adaptive immunity, which is further enhanced by blockade of PD-1/PD-L1 pathway, thereby providing a rationale for combining MDM2 inhibitors and checkpoint blocking antibodies in patients with wild-type p53 tumors. SIGNIFICANCE: This study provides a mechanistic rationale for combining checkpoint blockade immunotherapy with MDM2 inhibitors in patients with wild-type p53 tumors.


Sujet(s)
Lymphocytes T CD8+/immunologie , Tumeurs du côlon/traitement médicamenteux , Régulation de l'expression des gènes tumoraux , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Cellules stromales/immunologie , Microenvironnement tumoral/immunologie , Protéine p53 suppresseur de tumeur/antagonistes et inhibiteurs , Animaux , Apoptose , Prolifération cellulaire , Tumeurs du côlon/immunologie , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Association de médicaments , Femelle , Humains , Imidazoles/pharmacologie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris de lignée DBA , Souris nude , Pyrimidines/pharmacologie , Pyrroles/pharmacologie , Cellules stromales/effets des médicaments et des substances chimiques , Cellules cancéreuses en culture , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Cancer Cell ; 38(6): 872-890.e6, 2020 12 14.
Article de Anglais | MEDLINE | ID: mdl-33217342

RÉSUMÉ

Acquired resistance to BH3 mimetic antagonists of BCL-2 and MCL-1 is an important clinical problem. Using acute myelogenous leukemia (AML) patient-derived xenograft (PDX) models of acquired resistance to BCL-2 (venetoclax) and MCL-1 (S63845) antagonists, we identify common principles of resistance and persistent vulnerabilities to overcome resistance. BH3 mimetic resistance is characterized by decreased mitochondrial apoptotic priming as measured by BH3 profiling, both in PDX models and human clinical samples, due to alterations in BCL-2 family proteins that vary among cases, but not to acquired mutations in leukemia genes. BCL-2 inhibition drives sequestered pro-apoptotic proteins to MCL-1 and vice versa, explaining why in vivo combinations of BCL-2 and MCL-1 antagonists are more effective when concurrent rather than sequential. Finally, drug-induced mitochondrial priming measured by dynamic BH3 profiling (DBP) identifies drugs that are persistently active in BH3 mimetic-resistant myeloblasts, including FLT-3 inhibitors and SMAC mimetics.


Sujet(s)
Composés hétérocycliques bicycliques/pharmacologie , Résistance aux médicaments antinéoplasiques , Leucémie aigüe myéloïde/anatomopathologie , Mitochondries/métabolisme , Pyrimidines/pharmacologie , Sulfonamides/pharmacologie , Thiophènes/pharmacologie , Animaux , Apoptose , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/métabolisme , Souris , Mitochondries/effets des médicaments et des substances chimiques , Fragments peptidiques/pharmacologie , Protéines proto-oncogènes/pharmacologie , Transduction du signal
9.
Breast Cancer Res ; 22(1): 87, 2020 08 12.
Article de Anglais | MEDLINE | ID: mdl-32787886

RÉSUMÉ

BACKGROUND: Resistance to endocrine therapy is a major clinical challenge in the management of oestrogen receptor (ER)-positive breast cancer. In this setting, p53 is frequently wildtype and its activity may be suppressed via upregulation of its key regulator MDM2. This underlies our rationale to evaluate MDM2 inhibition as a therapeutic strategy in treatment-resistant ER-positive breast cancer. METHODS: We used the MDM2 inhibitor NVP-CGM097 to treat in vitro and in vivo models alone and in combination with fulvestrant or palbociclib. We perform cell viability, cell cycle, apoptosis and senescence assays to evaluate anti-tumour effects in p53 wildtype and p53 mutant ER-positive cell lines (MCF-7, ZR75-1, T-47D) and MCF-7 lines resistant to endocrine therapy and to CDK4/6 inhibition. We further assess the drug effects in patient-derived xenograft (PDX) models of endocrine-sensitive and endocrine-resistant ER-positive breast cancer. RESULTS: We demonstrate that MDM2 inhibition results in cell cycle arrest and increased apoptosis in p53-wildtype in vitro and in vivo breast cancer models, leading to potent anti-tumour activity. We find that endocrine therapy or CDK4/6 inhibition synergises with MDM2 inhibition but does not further enhance apoptosis. Instead, combination treatments result in profound regulation of cell cycle-related transcriptional programmes, with synergy achieved through increased antagonism of cell cycle progression. Combination therapy pushes cell lines resistant to fulvestrant or palbociclib to become senescent and significantly reduces tumour growth in a fulvestrant-resistant patient-derived xenograft model. CONCLUSIONS: We conclude that MDM2 inhibitors in combination with ER degraders or CDK4/6 inhibitors represent a rational strategy for treating advanced, endocrine-resistant ER-positive breast cancer, operating through synergistic activation of cell cycle co-regulatory programmes.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Tumeurs du sein/traitement médicamenteux , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Résistance aux médicaments antinéoplasiques , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Récepteurs des oestrogènes/métabolisme , Animaux , Apoptose , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire , Femelle , Fulvestrant/administration et posologie , Humains , Isoquinoléines/administration et posologie , Souris , Souris de lignée NOD , Souris SCID , Pipérazines/administration et posologie , Pyridines/administration et posologie , Tests d'activité antitumorale sur modèle de xénogreffe
10.
Eur J Cancer ; 126: 93-103, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31927215

RÉSUMÉ

INTRODUCTION: Uveal melanoma (UM) is a rare and malignant intraocular tumour with a dismal prognosis. Despite a good control of the primary tumour by radiation or surgery, up to 50% of patients subsequently develop metastasis for which no efficient treatment is yet available. METHODOLOGY: To identify therapeutic opportunities, we performed an in vitro screen of 30 combinations of different inhibitors of pathways that are dysregulated in UM. Effects of drug combinations on viability, cell cycle and apoptosis were assessed in eight UM cell lines. The best synergistic combinations were further evaluated in six UM patient-derived xenografts (PDXs). RESULTS: We demonstrated that the Bcl-2/XL/W inhibitor (ABT263) sensitised the UM cell lines to other inhibitors, mainly to mammalian target of rapamycin (mTOR), mitogen-activated protein kinase kinase (MEK) and murine double minute 2 (MDM2) inhibitors. mTOR (RAD001) and MEK1/2 (trametinib) inhibitors were efficient as single agents, but their combinations with ABT263 displayed no synergism in UM PDXs. In contrast, the combination of ABT263 with MDM2 inhibitor (HDM201) showed a trend for a synergistic effect. CONCLUSION: We showed that inhibition of Bcl-2/XL/W sensitised the UM cell lines to other treatments encouraging investigation of the underlying mechanisms. Furthermore, our findings highlighted Bcl-2/XL/W and MDM2 co-inhibition as a promising strategy in UM.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Évaluation préclinique de médicament/méthodes , Mélanome/traitement médicamenteux , Tumeurs de l'uvée/traitement médicamenteux , Dérivés de l'aniline/administration et posologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Protéines régulatrices de l'apoptose/antagonistes et inhibiteurs , Protéines régulatrices de l'apoptose/métabolisme , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Association médicamenteuse , Évérolimus/administration et posologie , Humains , Imidazoles/administration et posologie , Mélanome/métabolisme , Mélanome/anatomopathologie , Souris , Mitogen-Activated Protein Kinase Kinases/antagonistes et inhibiteurs , Mitogen-Activated Protein Kinase Kinases/métabolisme , Protéines proto-oncogènes c-bcl-2/antagonistes et inhibiteurs , Protéines proto-oncogènes c-bcl-2/métabolisme , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Protéines proto-oncogènes c-mdm2/métabolisme , Pyridones/administration et posologie , Pyrimidines/administration et posologie , Pyrimidinones/administration et posologie , Pyrroles/administration et posologie , Sulfonamides/administration et posologie , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Sérine-thréonine kinases TOR/métabolisme , Tumeurs de l'uvée/métabolisme , Tumeurs de l'uvée/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes , Protéine bcl-X/antagonistes et inhibiteurs , Protéine bcl-X/métabolisme
12.
Sci Transl Med ; 11(505)2019 08 14.
Article de Anglais | MEDLINE | ID: mdl-31413145

RÉSUMÉ

Intrinsic resistance of unknown mechanism impedes the clinical utility of inhibitors of cyclin-dependent kinases 4 and 6 (CDK4/6i) in malignancies other than breast cancer. Here, we used melanoma patient-derived xenografts (PDXs) to study the mechanisms for CDK4/6i resistance in preclinical settings. We observed that melanoma PDXs resistant to CDK4/6i frequently displayed activation of the phosphatidylinositol 3-kinase (PI3K)-AKT pathway, and inhibition of this pathway improved CDK4/6i response in a p21-dependent manner. We showed that a target of p21, CDK2, was necessary for proliferation in CDK4/6i-treated cells. Upon treatment with CDK4/6i, melanoma cells up-regulated cyclin D1, which sequestered p21 and another CDK inhibitor, p27, leaving a shortage of p21 and p27 available to bind and inhibit CDK2. Therefore, we tested whether induction of p21 in resistant melanoma cells would render them responsive to CDK4/6i. Because p21 is transcriptionally driven by p53, we coadministered CDK4/6i with a murine double minute (MDM2) antagonist to stabilize p53, allowing p21 accumulation. This resulted in improved antitumor activity in PDXs and in murine melanoma. Furthermore, coadministration of CDK4/6 and MDM2 antagonists with standard of care therapy caused tumor regression. Notably, the molecular features associated with response to CDK4/6 and MDM2 inhibitors in PDXs were recapitulated by an ex vivo organotypic slice culture assay, which could potentially be adopted in the clinic for patient stratification. Our findings provide a rationale for cotargeting CDK4/6 and MDM2 in melanoma.


Sujet(s)
Kinase-4 cycline-dépendante/métabolisme , Kinase-6 cycline-dépendante/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Protéines proto-oncogènes c-mdm2/métabolisme , Analyse de variance , Animaux , Technique de Western , Cycle cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Kinase-4 cycline-dépendante/génétique , Kinase-6 cycline-dépendante/génétique , Inhibiteur p21 de kinase cycline-dépendante/génétique , Réplication de l'ADN/effets des médicaments et des substances chimiques , Réplication de l'ADN/génétique , Diméthylsulfoxyde/pharmacologie , Humains , Immunoprécipitation , Cellules MCF-7 , Mélanome/métabolisme , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris nude , Protéomique , Dosage par radioimmunoprécipitation
13.
Leukemia ; 33(4): 905-917, 2019 04.
Article de Anglais | MEDLINE | ID: mdl-30214012

RÉSUMÉ

Improving outcomes in acute myeloid leukemia (AML) remains a major clinical challenge. Overexpression of pro-survival BCL-2 family members rendering transformed cells resistant to cytotoxic drugs is a common theme in cancer. Targeting BCL-2 with the BH3-mimetic venetoclax is active in AML when combined with low-dose chemotherapy or hypomethylating agents. We now report the pre-clinical anti-leukemic efficacy of a novel BCL-2 inhibitor S55746, which demonstrates synergistic pro-apoptotic activity in combination with the MCL1 inhibitor S63845. Activity of the combination was caspase and BAX/BAK dependent, superior to combination with standard cytotoxic AML drugs and active against a broad spectrum of poor risk genotypes, including primary samples from patients with chemoresistant AML. Co-targeting BCL-2 and MCL1 was more effective against leukemic, compared to normal hematopoietic progenitors, suggesting a therapeutic window of activity. Finally, S55746 combined with S63845 prolonged survival in xenograft models of AML and suppressed patient-derived leukemia but not normal hematopoietic cells in bone marrow of engrafted mice. In conclusion, a dual BH3-mimetic approach is feasible, highly synergistic, and active in diverse models of human AML. This approach has strong clinical potential to rapidly suppress leukemia, with reduced toxicity to normal hematopoietic precursors compared to chemotherapy.


Sujet(s)
Biomimétique , Composés hétérocycliques bicycliques/pharmacologie , Leucémie aigüe myéloïde/traitement médicamenteux , Protéine Mcl-1/antagonistes et inhibiteurs , Protéines proto-oncogènes c-bcl-2/antagonistes et inhibiteurs , Pyrimidines/pharmacologie , Sulfonamides/pharmacologie , Thiophènes/pharmacologie , Animaux , Antinéoplasiques/pharmacologie , Association de médicaments , Femelle , Humains , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Mâle , Souris , Souris de lignée NOD , Souris SCID , Fragments peptidiques , Protéines proto-oncogènes , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
14.
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
15.
Neuroendocrinology ; 106(1): 1-19, 2018.
Article de Anglais | MEDLINE | ID: mdl-27871087

RÉSUMÉ

BACKGROUND/AIMS: The tumor suppressor p53 is depleted in many tumor cells by the E3 ubiquitin ligase mouse double minute 2 homolog (MDM2) through MDM2/p53 interaction. A novel target for inhibiting p53 degradation and for causing reexpression of p53wild type is inhibition of MDM2. The small molecule NVP-CGM097 is a novel MDM2 inhibitor. We investigated MDM2 inhibition as a target in neuroendocrine tumor cells in vitro. METHODS: Human neuroendocrine tumor cell lines from the pancreas (BON1), lung (NCI-H727), and midgut (GOT1) were incubated with the MDM2 inhibitor NVP-CGM097 (Novartis) at concentrations from 4 to 2,500 nM. RESULTS: While p53wild type GOT1 cells were sensitive to NVP-CGM097, p53mutated BON1 and p53mutated NCI-H727 cells were resistant to NVP-CGM097. Incubation of GOT1 cells with NVP-CGM097 at 100, 500, and 2,500 nM for 96 h caused a significant decline in cell viability to 84.9 ± 9.2% (p < 0.05), 77.4 ± 6.6% (p < 0.01), and 47.7 ± 9.2% (p < 0.01). In a Western blot analysis of GOT1 cells, NVP-CGM097 caused a dose-dependent increase in the expression of p53 and p21 tumor suppressor proteins and a decrease in phospho-Rb and E2F1. Experiments of co-incubation of NVP-CGM097 with 5-fluorouracil, temozolomide, or everolimus each showed additive antiproliferative effects in GOT1 cells. NVP-CGM097 and 5-fluorouracil increased p53 and p21 expression in an additive manner. CONCLUSIONS: MDM2 inhibition seems a promising novel therapeutic target in neuroendocrine tumors harboring p53wild type. Further investigations should examine the potential role of MDM2 inhibitors in neuroendocrine tumor treatment.


Sujet(s)
Antinéoplasiques/pharmacologie , Fluorouracil/pharmacologie , Isoquinoléines/pharmacologie , Tumeurs neuroendocrines/traitement médicamenteux , Pipérazines/pharmacologie , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Relation dose-effet des médicaments , Résistance aux médicaments antinéoplasiques , Association de médicaments , Facteur de transcription E2F1/métabolisme , Humains , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/métabolisme , Protéines proto-oncogènes c-mdm2/métabolisme , Protéine du rétinoblastome/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/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.
Elife ; 62017 04 20.
Article de Anglais | MEDLINE | ID: mdl-28425916

RÉSUMÉ

The efficacy of ALK inhibitors in patients with ALK-mutant neuroblastoma is limited, highlighting the need to improve their effectiveness in these patients. To this end, we sought to develop a combination strategy to enhance the antitumor activity of ALK inhibitor monotherapy in human neuroblastoma cell lines and xenograft models expressing activated ALK. Herein, we report that combined inhibition of ALK and MDM2 induced a complementary set of anti-proliferative and pro-apoptotic proteins. Consequently, this combination treatment synergistically inhibited proliferation of TP53 wild-type neuroblastoma cells harboring ALK amplification or mutations in vitro, and resulted in complete and durable responses in neuroblastoma xenografts derived from these cells. We further demonstrate that concurrent inhibition of MDM2 and ALK was able to overcome ceritinib resistance conferred by MYCN upregulation in vitro and in vivo. Together, combined inhibition of ALK and MDM2 may provide an effective treatment for TP53 wild-type neuroblastoma with ALK aberrations.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Résistance aux médicaments antinéoplasiques , Neuroblastome/traitement médicamenteux , Neuroblastome/anatomopathologie , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Kinase du lymphome anaplasique , Animaux , Antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Hétérogreffes , Humains , Souris nude , Transplantation tumorale , Pyrimidines/pharmacologie , Pyrimidines/usage thérapeutique , Sulfones/pharmacologie , Sulfones/usage thérapeutique
19.
Cancer Res ; 76(23): 6950-6963, 2016 12 01.
Article de Anglais | MEDLINE | ID: mdl-27659046

RÉSUMÉ

Like classical chemotherapy regimens used to treat cancer, targeted therapies will also rely upon polypharmacology, but tools are still lacking to predict which combinations of molecularly targeted drugs may be most efficacious. In this study, we used image-based proliferation and apoptosis assays in colorectal cancer cell lines to systematically investigate the efficacy of combinations of two to six drugs that target critical oncogenic pathways. Drug pairs targeting key signaling pathways resulted in synergies across a broad spectrum of genetic backgrounds but often yielded only cytostatic responses. Enhanced cytotoxicity was observed when additional processes including apoptosis and cell cycle were targeted as part of the combination. In some cases, where cell lines were resistant to paired and tripled drugs, increased expression of antiapoptotic proteins was observed, requiring a fourth-order combination to induce cytotoxicity. Our results illustrate how high-order drug combinations are needed to kill drug-resistant cancer cells, and they also show how systematic drug combination screening together with a molecular understanding of drug responses may help define optimal cocktails to overcome aggressive cancers. Cancer Res; 76(23); 6950-63. ©2016 AACR.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs colorectales/traitement médicamenteux , Animaux , Prolifération cellulaire , Tumeurs colorectales/génétique , Femelle , Humains , Souris , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE