Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 27
Filtrer
1.
J Immunother Cancer ; 11(11)2023 11 29.
Article de Anglais | MEDLINE | ID: mdl-38030303

RÉSUMÉ

BACKGROUND: NIS793 is a human IgG2 monoclonal antibody that binds to transforming growth factor beta (TGF-ß). This first-in-human study investigated NIS793 plus spartalizumab treatment in patients with advanced solid tumors. METHODS: Patients received NIS793 (0.3-1 mg/kg every 3 weeks (Q3W)) monotherapy; following evaluation of two dose levels, dose escalation continued with NIS793 plus spartalizumab (NIS793 0.3-30 mg/kg Q3W and spartalizumab 300 mg Q3W or NIS793 20-30 mg/kg every 2 weeks [Q2W] and spartalizumab 400 mg every 4 weeks (Q4W)). In dose expansion, patients with non-small cell lung cancer (NSCLC) resistant to prior anti-programmed death ligand 1 or patients with microsatellite stable colorectal cancer (MSS-CRC) were treated at the recommended dose for expansion (RDE). RESULTS: Sixty patients were treated in dose escalation, 11 with NIS793 monotherapy and 49 with NIS793 plus spartalizumab, and 60 patients were treated in dose expansion (MSS-CRC: n=40; NSCLC: n=20). No dose-limiting toxicities were observed. The RDE was established as NIS793 30 mg/kg (2100 mg) and spartalizumab 300 mg Q3W. Overall 54 (49.5%) patients experienced ≥1 treatment-related adverse event, most commonly rash (n=16; 13.3%), pruritus (n=10; 8.3%), and fatigue (n=9; 7.5%). Three partial responses were reported: one in renal cell carcinoma (NIS793 30 mg/kg Q2W plus spartalizumab 400 mg Q4W), and two in the MSS-CRC expansion cohort. Biomarker data showed evidence of target engagement through increased TGF-ß/NIS793 complexes and depleted active TGF-ß in peripheral blood. Gene expression analyses in tumor biopsies demonstrated decreased TGF-ß target genes and signatures and increased immune signatures. CONCLUSIONS: In patients with advanced solid tumors, proof of mechanism of NIS793 is supported by evidence of target engagement and TGF-ß pathway inhibition. TRIAL REGISTRATION NUMBER: NCT02947165.


Sujet(s)
Antinéoplasiques , Carcinome pulmonaire non à petites cellules , Tumeurs du rein , Tumeurs du poumon , Adulte , Humains , Anticorps monoclonaux/effets indésirables , Antinéoplasiques/usage thérapeutique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du rein/traitement médicamenteux , Tumeurs du poumon/traitement médicamenteux , Facteur de croissance transformant bêta
3.
Mol Cancer Ther ; 21(4): 625-634, 2022 04 01.
Article de Anglais | MEDLINE | ID: mdl-35131875

RÉSUMÉ

This first-in-human (FIH), phase I, multicenter, open-label study was conducted to characterize the safety, tolerability, pharmacokinetics, and preliminary efficacy, and to establish the MTD/recommended dose for expansion (RDE) of PCA062 in patients with solid tumors. Adult patients with any solid tumor type and having a documented P-cadherin-positive tumor were enrolled; exceptions to P-cadherin positivity requirement were head and neck squamous cell carcinomas (HNSCC) and esophageal squamous cell carcinoma (ESCC). Dose escalation was guided by an adaptive Bayesian logistic regression model with escalation with overdose control to determine the MTD/RDE. Forty-seven patients were treated at 10 different dose levels of PCA062, ranging from 0.4 to 5.0 mg/kg every 2 weeks administered as a 1-hour intravenous infusion. All enrolled patients discontinued the treatment; primary reason for discontinuation was progressive disease (78.7%). All 47 patients experienced at least one AE, of which 32 patients had a grade ≥3 AE and 37 patients experienced AEs suspected to be study drug related. The MTD of PCA062 was 3.6 mg/kg every 2 weeks and thrombocytopenia was reported as a DLT that was attributed to the known toxicities of the DM1 payload with no P-cadherin-related toxicities. Pharmacokinetics was proportional, and no patients developed antidrug antibodies, suggesting adequate exposure at the doses tested. One patient of 47 achieved a partial response and there was no correlation between tumor P-cadherin expression and clinical efficacy. Because of limited antitumor activity at the MTD level, Novartis has terminated clinical development of PCA062 (NCT02375958).


Sujet(s)
Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Tumeurs de la tête et du cou , Immunoconjugués , Tumeurs , Adulte , Théorème de Bayes , Cadhérines , Tumeurs de l'oesophage/traitement médicamenteux , Tumeurs de la tête et du cou/traitement médicamenteux , Humains , Immunoconjugués/usage thérapeutique , Dose maximale tolérée , Tumeurs/anatomopathologie
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.
AAPS J ; 23(2): 28, 2021 02 07.
Article de Anglais | MEDLINE | ID: mdl-33554304

RÉSUMÉ

We report on a retrospective model-based assessment of the predictive value of translating antitumor drug activity from in vivo experiments to a phase I clinical study in cancer patients treated with the MDM2 inhibitor, HDM201. Tumor growth inhibition models were developed describing the longitudinal tumor size data in human-derived osteosarcoma xenograft rats and in 96 solid tumor patients under different HDM201 treatment schedules. The model structure describing both datasets captures the delayed drug effect on tumor growth via a series of signal transduction compartments, including a resistance component. The models assumed a drug-killing effect on both sensitive and resistant cells and parameterized to estimate two tumor static plasma drug concentrations for sensitive (TSCS) and resistant cells (TSCR). No change of TSCS and TSCR with schedule was observed, implying that antitumor activity for HDM201 is independent of treatment schedule. Preclinical and clinical model-derived TSCR were comparable (48 ng/mL vs. 74 ng/mL) and demonstrating TSCR as a translatable metric for antitumor activity in clinic. Schedule independency was further substantiated from modeling of clinical serum growth differentiation factor-15 (GDF-15) as a downstream marker of p53 pathway activation. Equivalent cumulative induction of GDF-15 was achieved across schedules when normalized to an equivalent total dose. These findings allow for evaluation of optimal dosing schedules by maximizing the total dose per treatment cycle while mitigating safety risk with periods of drug holiday. This approach helped guide a phase I dose escalation study in the selection of an optimal dose and schedule for HDM201.


Sujet(s)
Imidazoles/administration et posologie , Modèles biologiques , Ostéosarcome/traitement médicamenteux , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Pyrimidines/administration et posologie , Pyrroles/administration et posologie , Administration par voie orale , Adolescent , Adulte , Animaux , Biodisponibilité , Lignée cellulaire tumorale , Relation dose-effet des médicaments , Calendrier d'administration des médicaments , Résistance aux médicaments antinéoplasiques , Femelle , Facteur-15 de croissance et de différenciation/sang , Facteur-15 de croissance et de différenciation/métabolisme , Humains , Imidazoles/effets indésirables , Imidazoles/pharmacocinétique , Mâle , Adulte d'âge moyen , Ostéosarcome/sang , Ostéosarcome/génétique , Pyrimidines/effets indésirables , Pyrimidines/pharmacocinétique , Pyrroles/effets indésirables , Pyrroles/pharmacocinétique , Rats , Évaluation de la réponse des tumeurs solides aux traitements , Études rétrospectives , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe , Jeune adulte
8.
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
9.
Nat Commun ; 11(1): 6315, 2020 12 09.
Article de Anglais | MEDLINE | ID: mdl-33298926

RÉSUMÉ

Despite the increasing interest in targeting stromal elements of the tumor microenvironment, we still face tremendous challenges in developing adequate therapeutics to modify the tumor stromal landscape. A major obstacle to this is our poor understanding of the phenotypic and functional heterogeneity of stromal cells in tumors. Herein, we perform an unbiased interrogation of tumor mesenchymal cells, delineating the co-existence of distinct subsets of cancer-associated fibroblasts (CAFs) in the microenvironment of murine carcinomas, each endowed with unique phenotypic features and functions. Furthermore, our study shows that neutralization of TGFß in vivo leads to remodeling of CAF dynamics, greatly reducing the frequency and activity of the myofibroblast subset, while promoting the formation of a fibroblast population characterized by strong response to interferon and heightened immunomodulatory properties. These changes correlate with the development of productive anti-tumor immunity and greater efficacy of PD1 immunotherapy. Along with providing the scientific rationale for the evaluation of TGFß and PD1 co-blockade in the clinical setting, this study also supports the concept of plasticity of the stromal cell landscape in tumors, laying the foundation for future investigations aimed at defining pathways and molecules to program CAF composition for cancer therapy.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Fibroblastes associés au cancer/immunologie , Carcinomes/traitement médicamenteux , Interféron bêta/immunologie , Facteur de croissance transformant bêta/antagonistes et inhibiteurs , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques , Carcinomes/immunologie , Carcinomes/anatomopathologie , Lignée cellulaire tumorale/transplantation , Plasticité cellulaire/effets des médicaments et des substances chimiques , Plasticité cellulaire/immunologie , Modèles animaux de maladie humaine , Synergie des médicaments , Femelle , Humains , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Souris , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Cellules stromales/effets des médicaments et des substances chimiques , Cellules stromales/immunologie , Facteur de croissance transformant bêta/métabolisme , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie
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.
Invest New Drugs ; 37(2): 271-281, 2019 04.
Article de Anglais | MEDLINE | ID: mdl-30073466

RÉSUMÉ

Background CLR457 is an orally bioavailable pan-phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) inhibitor. Methods CLR457 anti-tumor activity and pharmacokinetics (PK) were characterized by in vitro biochemical assays and in vivo tumor xenografts. A first-in-human study was conducted to determine the maximum tolerated dose (MTD), safety, PK, and efficacy of CLR457. Successive cohorts of patients with advanced solid tumors with PI3K pathway activation received increasing CLR457 doses according to a Bayesian escalation model based on the rate of dose limiting toxicity (DLT) in the first 28-day cycle. Results CLR457 inhibited p110α, p110ß, p110δ and p110γ isoforms with an IC50 of 89 ± 29 nM, 56 ± 35 nM, 39 ± 10 nM and 230 ± 31 nM, respectively. CLR457 exhibited dose-dependent antitumor activity and interfered with glucose homeostasis in PI3K-mutant tumor xenografts. 31 patients received doses ranging from 5 to 100 mg. DLTs included grade 3 hyperglycemia and rash (3). In the 100 mg cohort (n = 11), 3 (27.3%) patients had DLTs and all patients (100%) experienced ≥ grade 3 toxicity with rash (45.5%) as the most common event. The MTD was not determined. For the entire study population, stomatitis (45.2%), diarrhea (38.7%), rash (35.5%) were the most common any grade toxicities-51.6% patients experienced ≥ Grade 3 toxicity. CLR457 was rapidly absorbed with limited accumulation and linear PK. PK modeling indicated that pharmacologically active concentrations were achieved at the highest dose tested (100 mg), though no objective responses were observed. Conclusion CLR457 clinical development was terminated due to poor tolerability and limited antitumor activity. These results emphasize the difficulty of achieving a wide therapeutic index when targeting all class I PI3K-isoforms.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Phosphatidylinositol 3-kinases de classe I/antagonistes et inhibiteurs , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Composés chimiques organiques/administration et posologie , Inhibiteurs de protéines kinases/administration et posologie , Administration par voie orale , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Apoptose , Prolifération cellulaire , Relation dose-effet des médicaments , Femelle , Études de suivi , Humains , Mâle , Dose maximale tolérée , Souris , Souris nude , Adulte d'âge moyen , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Pronostic , Rat nude , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
14.
Cancer Chemother Pharmacol ; 74(5): 1057-64, 2014 Nov.
Article de Anglais | MEDLINE | ID: mdl-25217392

RÉSUMÉ

PURPOSE: Dinaciclib is a novel selective inhibitor of cyclin-dependent kinase (CDK)1, CDK2, CDK5, and CDK9. We conducted a phase I study to investigate the effects of dinaciclib when administered with rituximab. METHODS: In this phase I nonrandomized dose-escalation 3 + 3 trial, patients with relapsed/refractory chronic lymphocytic leukemia (CLL) were treated with dinaciclib and rituximab. Dinaciclib was administered intravenously (IV) over 2 h on days 1, 8 and 15 in cycles 2-13 (28-day cycles). Rituximab 375 mg/m(2) was administered IV on days 1, 8, 15 and 22 in cycle 1 (28-day cycle) and on day 1 during cycle 3-13. Rituximab was not administered in cycle 2. Rituximab and dinaciclib were given alone in cycles 1 and 2, respectively, and in combination in cycles 3-13. Primary objectives included determination of the recommended phase II dose of dinaciclib and evaluation of pharmacokinetics (PK) when administered with rituximab. RESULTS: Five patients completed the study due to early termination. All presented with drug-related adverse events (AEs), but no dose-limiting toxicities were observed. The most commonly observed toxicities included hematological, digestive and metabolic AEs. However, no tumor lysis syndrome has been reported in the study. Four patients achieved stable disease, and one patient achieved complete response according to 2008 iwCLL criteria at cycle 3. PK samples were collected from 5 patients, and no obvious interaction between dinaciclib and rituximab was observed. CONCLUSIONS: Limited data from this study shows dinaciclib in combination with rituximab was well tolerated and revealed encouraging clinical activity in relapsed/refractory CLL patients.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Kinases cyclines-dépendantes/antagonistes et inhibiteurs , Leucémie chronique lymphocytaire à cellules B/traitement médicamenteux , Administration par voie intraveineuse , Sujet âgé , Anémie/induit chimiquement , Anticorps monoclonaux d'origine murine/administration et posologie , Anticorps monoclonaux d'origine murine/effets indésirables , Anticorps monoclonaux d'origine murine/pharmacocinétique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/pharmacocinétique , Aire sous la courbe , Asthénie/induit chimiquement , Composés hétérocycliques bicycliques/administration et posologie , Composés hétérocycliques bicycliques/effets indésirables , Composés hétérocycliques bicycliques/pharmacocinétique , N-oxydes cycliques , Kinases cyclines-dépendantes/métabolisme , Diarrhée/induit chimiquement , Calendrier d'administration des médicaments , Résistance aux médicaments antinéoplasiques , Femelle , Humains , Indolizine , Leucémie chronique lymphocytaire à cellules B/métabolisme , Leucémie chronique lymphocytaire à cellules B/anatomopathologie , Mâle , Taux de clairance métabolique , Adulte d'âge moyen , Récidive tumorale locale , Composés de pyridinium/administration et posologie , Composés de pyridinium/effets indésirables , Composés de pyridinium/pharmacocinétique , Rituximab , Résultat thérapeutique
15.
Clin Lymphoma Myeloma Leuk ; 13(4): 370-6, 2013 Aug.
Article de Anglais | MEDLINE | ID: mdl-23787122

RÉSUMÉ

Multiple myeloma (MM) arises from abnormal proliferation and survival (ie, a high proliferative index and a low apoptotic index) of mature immunoglobulin-producing plasma cells in the bone marrow. Development of novel therapeutic options, such as proteasome inhibitors and immunomodulatory agents (IMiDs), has improved treatment outcomes. However, patients often develop relapsed and refractory MM, thus requiring alternative treatment approaches. Histone acetyltransferases and histone deacetylases (HDACs) control the acetylation status of proteins and affect a broad array of physiologic processes (eg, cell cycle, apoptosis, and protein folding) involved in cell growth and survival. The discovery that HDACs might have a role in various hematologic malignancies, including MM, has led to the development of HDAC inhibitors as potential antitumor agents. Preclinical evidence from studies of HDAC inhibitors in combination with proteasome inhibitors (eg, bortezomib and carfilzomib), other antimyeloma agents, including IMiDs (eg, lenalidomide), and cytotoxic agents (eg, melphalan, pegylated liposomal doxorubicin), provides a strong scientific rationale for the evaluation of these regimens. Results from early stage clinical trials further support the use of HDAC inhibitors as a therapeutic option for MM, in combination with current and emerging antimyeloma agents. In this review, we examine the role of protein acetylation that underlies the antimyeloma effects of HDAC inhibitors, discuss the preclinical rationale for the use of HDAC inhibitors in combination with other antimyeloma agents, and provide an overview of the current clinical evidence supporting the use of HDAC inhibitors as a therapeutic option in MM.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Inhibiteurs de désacétylase d'histone/administration et posologie , Myélome multiple/traitement médicamenteux , Humains , Myélome multiple/enzymologie , Myélome multiple/anatomopathologie
16.
PLoS One ; 8(3): e59127, 2013.
Article de Anglais | MEDLINE | ID: mdl-23555623

RÉSUMÉ

The NF-κB family of transcription factors has emerged as a key player in the pathogenesis of multiple myeloma (MM). NF-κB is activated by at least two major signaling pathways. The classical pathway results in the activation of mainly RelA containing dimers, whereas the alternative pathway leads to the activation of RelB/p52 and RelB/p50 heterodimers. Activating mutations in regulators of the alternative pathway have been identified in 17% of MM patients. However, the status of RelB activation per se and its role in the regulation of cell survival in MM has not been investigated. Here, we reveal that 40% of newly diagnosed MM patients have a constitutive RelB DNA-binding activity in CD138(+) tumor cells, and we show an association with increased expression of a subset of anti-apoptotic NF-κB target genes, such as cIAP2. Furthermore, we demonstrate that RelB exerts a crucial anti-apoptotic activity in MM cells. Our findings indicate that RelB activation is key for promoting MM cell survival through the upregulation of anti-apoptotic proteins. Altogether, our study provides the framework for the development of new molecules targeting RelB in the treatment of MM.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Protéines IAP/génétique , Myélome multiple/génétique , Facteur de transcription RelB/génétique , Transcription génétique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Apoptose , Protéine-3 contenant des répétitions IAP baculovirales , Survie cellulaire , ADN/métabolisme , Femelle , Humains , Protéines IAP/métabolisme , Mâle , Adulte d'âge moyen , Myélome multiple/métabolisme , Myélome multiple/anatomopathologie , Culture de cellules primaires , Transduction du signal , Syndécane-1/génétique , Syndécane-1/métabolisme , Facteur de transcription RelA/génétique , Facteur de transcription RelA/métabolisme , Facteur de transcription RelB/métabolisme , Ubiquitin-protein ligases
17.
Curr Hematol Malig Rep ; 7(4): 258-66, 2012 Dec.
Article de Anglais | MEDLINE | ID: mdl-23065395

RÉSUMÉ

Proteasome inhibition has a validated role in cancer therapy since the successful introduction of bortezomib for the treatment of multiple myeloma (MM) and mantle cell lymphoma, leading to the development of second-generation proteasome inhibitors (PI) for MM patients in whom currently approved therapies have failed. Five PIs have reached clinical evaluation, with the goals of improving efficacy and limiting toxicity, including peripheral neuropathy (PN). Carfilzomib, an epoxyketone with specific chymothrypsin-like activity, acts as an irreversible inhibitor and was recently FDA approved for the response benefit seen in relapsed and refractory MM patients previously treated with bortezomib, thalidomide and lenalidomide. ONX-0912 is now under evaluation as an oral form with similar activity. The boronate peptides MLN9708 and CEP-18770 are orally bioactive bortezomib analogs with prolonged activity and greater tissue penetration. NPI-0052 (marizomib) is a unique, beta-lactone non-selective PI that has been shown to potently overcome bortezomib resistance in vitro. All of these second-generation PIs demonstrate encouraging anti-MM activity and appear to reduce the incidence of PN, with clinical trials ongoing.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Myélome multiple/traitement médicamenteux , Inhibiteurs du protéasome/usage thérapeutique , Essais cliniques comme sujet , Évaluation préclinique de médicament , Humains , Proteasome endopeptidase complex/composition chimique , Proteasome endopeptidase complex/physiologie
18.
Clin Cancer Res ; 18(17): 4669-81, 2012 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-22806876

RÉSUMÉ

PURPOSE: NF-κB transcription factor plays a key role in the pathogenesis of multiple myeloma in the context of the bone marrow microenvironment. Both canonical and noncanonical pathways contribute to total NF-κB activity. Recent studies have shown a critical role for the noncanonical pathway: selective inhibitors of the canonical pathway present a limited activity, mutations of the noncanonical pathway are frequent, and bortezomib-induced cytotoxicity cannot be fully attributed to inhibition of canonical NF-κB activity. EXPERIMENTAL DESIGN: Multiple myeloma cell lines, primary patient cells, and the human multiple myeloma xenograft murine model were used to examine the biologic impact of dual inhibition of both canonical and noncanonical NF-κB pathways. RESULTS: We show that PBS-1086 induces potent cytotoxicity in multiple myeloma cells but not in peripheral blood mononuclear cells. PBS-1086 overcomes the proliferative and antiapoptotic effects of the bone marrow milieu, associated with inhibition of NF-κB activity. Moreover, PBS-1086 strongly enhances the cytotoxicity of bortezomib in bortezomib-resistant multiple myeloma cell lines and patient multiple myeloma cells. PBS-1086 also inhibits osteoclastogenesis through an inhibition of RANK ligand (RANKL)-induced NF-κB activation. Finally, in a xenograft model of human multiple myeloma in the bone marrow milieu, PBS-1086 shows significant in vivo anti-multiple myeloma activity and prolongs host survival, associated with apoptosis and inhibition of both NF-κB pathways in tumor cells. CONCLUSIONS: Our data show that PBS-1086 is a promising dual inhibitor of the canonical and noncanonical NF-κB pathways. Our preclinical study therefore provides the framework for clinical evaluation of PBS-1086 in combination with bortezomib for the treatment of multiple myeloma and related bone lesions.


Sujet(s)
Cyclohexanones/administration et posologie , Composés époxy/administration et posologie , Myélome multiple , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Transduction du signal/effets des médicaments et des substances chimiques , Chlorométhyl cétones d'acides aminés/administration et posologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Acides boroniques/administration et posologie , Bortézomib , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Doxorubicine/administration et posologie , Humains , Agranulocytes/effets des médicaments et des substances chimiques , Souris , Myélome multiple/traitement médicamenteux , Myélome multiple/métabolisme , Myélome multiple/anatomopathologie , Facteur de transcription NF-kappa B/métabolisme , Ostéoclastes/cytologie , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoclastes/métabolisme , Pyrazines/administration et posologie , Ligand de RANK/métabolisme , Transplantation hétérologue
19.
Blood ; 119(24): 5772-81, 2012 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-22538852

RÉSUMÉ

Multiple myeloma (MM) cells are characterized by high protein synthesis resulting in chronic endoplasmic reticulum (ER) stress, which is adaptively managed by the unfolded protein response. Inositol-requiring enzyme 1α (IRE1α) is activated to splice X-box binding protein 1 (XBP1) mRNA, thereby increasing XBP1s protein, which in turn regulates genes responsible for protein folding and degradation during the unfolded protein response. In this study, we examined whether IRE1α-XBP1 pathway is a potential therapeutic target in MM using a small-molecule IRE1α endoribonuclease domain inhibitor MKC-3946. MKC-3946 triggered modest growth inhibition in MM cell lines, without toxicity in normal mononuclear cells. Importantly, it significantly enhanced cytotoxicity induced by bortezomib or 17-AAG, even in the presence of bone marrow stromal cells or exogenous IL-6. Both bortezomib and 17-AAG induced ER stress, evidenced by induction of XBP1s, which was blocked by MKC-3946. Apoptosis induced by these agents was enhanced by MKC-3946, associated with increased CHOP. Finally, MKC-3946 inhibited XBP1 splicing in a model of ER stress in vivo, associated with significant growth inhibition of MM cells. Taken together, our results demonstrate that blockade of XBP1 splicing by inhibition of IRE1α endoribonuclease domain is a potential therapeutic option in MM.


Sujet(s)
Protéines de liaison à l'ADN/génétique , Endoribonucleases/antagonistes et inhibiteurs , Myélome multiple/traitement médicamenteux , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Épissage des ARN/effets des médicaments et des substances chimiques , Facteurs de transcription/génétique , Animaux , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Benzoquinones/pharmacologie , Cellules de la moelle osseuse/cytologie , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/métabolisme , Acides boroniques/pharmacologie , Acides boroniques/usage thérapeutique , Bortézomib , Mort cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Endoribonucleases/métabolisme , Activation enzymatique/effets des médicaments et des substances chimiques , Antienzymes/composition chimique , Antienzymes/pharmacologie , Antienzymes/usage thérapeutique , Humains , Interleukine-6/pharmacologie , Lactames macrocycliques/pharmacologie , Souris , Myélome multiple/génétique , Myélome multiple/anatomopathologie , Protein-Serine-Threonine Kinases/métabolisme , Pyrazines/pharmacologie , Pyrazines/usage thérapeutique , Épissage des ARN/génétique , ARN messager/génétique , ARN messager/métabolisme , Facteurs de transcription des facteurs régulateurs X , Transduction du signal/effets des médicaments et des substances chimiques , Cellules stromales/cytologie , Cellules stromales/effets des médicaments et des substances chimiques , Cellules stromales/métabolisme , Réponse aux protéines mal repliées/effets des médicaments et des substances chimiques , Protéine-1 liant la boite X , eIF-2 Kinase/métabolisme
20.
Haematologica ; 97(4): 482-90, 2012 Apr.
Article de Anglais | MEDLINE | ID: mdl-22058218

RÉSUMÉ

BACKGROUND: How tandem autologous-allogeneic stem cell transplantation should be integrated in the treatment of multiple myeloma remains controversial. We examined the long-term outcome of patients with multiple myeloma managed with tandem autologous-allogeneic stem cell transplantation and present a prognostic factor analysis based on the experience of the Société Française de Greffe de Moelle et de Thérapie Cellulaire (SFGM-TC). DESIGN AND METHODS: This French, retrospective, registry-based study included 146 patients who had undergone tandem autologous-allogeneic transplantation for multiple myeloma at 20 SFGM-TC centers between 1998 and 2010. The patients included in the study had fully completed the two steps of a planned tandem autologous-allogeneic transplantation. No treatment had to be administered between the autologous and allogeneic parts of the tandem procedure. RESULTS: Seventy-seven patients (53%) underwent tandem autologous-allogeneic transplantation as part of upfront treatment, i.e. after a single line of treatment not including autologous transplantation. The median follow-up from the allogeneic transplant was 47.5 months (range, 1.2-132 months). At 4 years, the overall survival and event-free survival rates were 48% (95% CI 39-57 %) and 27% (95% CI 19-36), respectively. Eighteen patients (12%) experienced grade III-IV acute graft-versus-host disease and 43 patients (30%) had chronic graft-versus-host disease. The transplant-related mortality rate at 1 year was 15% (95% CI 10-22). Patients receiving tandem transplantation as upfront treatment had significantly improved event-free survival (36% versus 11%; P=0.005) and overall survival (56% versus 34%; P=0.02). Donor's age ≤ 50 years was associated with improved event-free survival (35% versus 16%; P=0.005) and overall survival (54% versus 41%; P=0.02). In the multivariable analysis, upfront tandem transplantation, donor's age ≤ 50 years and full chimerism were independent prognostic factors for better outcome. CONCLUSIONS: We confirmed the feasibility of tandem autologour-allogeneic transplantation in heavily treated patients with multiple myeloma. We identified younger donor's age and upfront tandem transplantation as two independent prognostic factors for survival which could be further explored in prospective studies.


Sujet(s)
Transplantation de cellules souches hématopoïétiques/mortalité , Myélome multiple/mortalité , Myélome multiple/thérapie , Donneurs de tissus , Adulte , Facteurs âges , Sujet âgé , Cause de décès , Femelle , Transplantation de cellules souches hématopoïétiques/effets indésirables , Humains , Mâle , Adulte d'âge moyen , Morbidité , Myélome multiple/épidémiologie , Pronostic , Récidive , Enregistrements , Études rétrospectives , Analyse de survie , Transplantation autologue , Transplantation homologue , Résultat thérapeutique , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...