22 research outputs found

    First-in-human Phase 1 open label study of the BET inhibitor ODM-207 in patients with selected solid tumours

    Get PDF
    Background Bromodomain and extra-terminal domain (BET) proteins are reported to be epigenetic anti-cancer drug targets. This first-in-human study evaluated the safety, pharmacokinetics and preliminary anti-tumour activity of the BET inhibitor ODM-207 in patients with selected solid tumours. Methods This was an open-label Phase 1 study comprised of a dose escalation part, and evaluation of the effect of food on pharmacokinetics. ODM-207 was administered orally once daily. The dose escalation part was initiated with a dose titration in the initial cohort, followed by a 3 + 3 design. Results Thirty-five patients were treated with ODM-207, of whom 12 (34%) had castrate-resistant prostate cancer. One dose-limiting toxicity of intolerable fatigue was observed. The highest studied dose achieved was 2 mg/kg due to cumulative toxicity observed beyond the dose-limiting toxicity (DLT) treatment window. Common AEs included thrombocytopenia, asthenia, nausea, anorexia, diarrhoea, fatigue, and vomiting. Platelet count decreased proportionally to exposure with rapid recovery upon treatment discontinuation. No partial or complete responses were observed. Conclusions ODM-207 shows increasing exposure in dose escalation and was safe at doses up to 2 mg/kg but had a narrow therapeutic window.Peer reviewe

    Evidence of pseudoprogression in patients treated with PD1/ PDL1 antibodies across tumor types

    Get PDF
    Background: PD(L)1 antibodies (anti-PD(L)-1) have been a major breakthrough in several types of cancer. Novel patterns of response and progression have been described with anti-PD(L)-1. We aimed at characterizing pseudoprogression (PSPD) among patients with various solid tumor types treated by anti-PD(L)-1. Methods: All consecutive patients (pts) enrolled in phase 1 trials with advanced solid tumors and lymphomas treated in phase I clinical trials evaluating monotherapy by anti-PD(L)-1 at Gustave Roussy were analyzed. We aimed to assess prevalence and outcome of PSPD across tumor types. We also intended to describe potential clinical and pathological factors associated with PSPD. Results: A total of 169 patients treated with anti-PD(L)-1 were included in the study. Most frequent tumor types included melanoma (n = 57) and non-small cell lung cancer (n = 19). At first tumor evaluation 77 patients (46%) presented with immune unconfirmed progressive disease. Six patients (8%) experienced PSPD: 2 patients with partial response; 4 patients with stable disease. Increase in target lesions in the first CT-scan was more frequently associated to PSPD (67% vs 33%; P = .04). Patients with a PSPD had a superior survival when compared to patients progressing (median OS: 10.7 months vs 8.7 months; P = .07). Conclusions: A small subset of PSPD patients may experience response after an initial progression. Assessment of the current strategy for immune-related response evaluations may require further attention

    Can molecular biomarker-based patient selection in Phase I trials accelerate anticancer drug development?

    No full text
    Anticancer drug development remains slow, costly and inefficient. One way of addressing this might be the use of predictive biomarkers to select patients for Phase I/II trials. Such biomarkers, which predict response to molecular-targeted agents, have the potential to enrich these trials with patients more likely to benefit. Doing so could maximize the efficiency of anticancer drug development by facilitating earlier clinical qualification of predictive biomarkers and generating valuable information on cancer biology. In this review, we suggest a new model of early clinical trial design, which incorporates patient selection through predictive molecular biomarkers for selected targeted agents

    Evidence of pseudoprogression in patients treated with PD1/ PDL1 antibodies across tumor types

    No full text
    Background: PD(L)1 antibodies (anti-PD(L)-1) have been a major breakthrough in several types of cancer. Novel patterns of response and progression have been described with anti-PD(L)-1. We aimed at characterizing pseudoprogression (PSPD) among patients with various solid tumor types treated by anti-PD(L)-1. Methods: All consecutive patients (pts) enrolled in phase 1 trials with advanced solid tumors and lymphomas treated in phase I clinical trials evaluating monotherapy by anti-PD(L)-1 at Gustave Roussy were analyzed. We aimed to assess prevalence and outcome of PSPD across tumor types. We also intended to describe potential clinical and pathological factors associated with PSPD. Results: A total of 169 patients treated with anti-PD(L)-1 were included in the study. Most frequent tumor types included melanoma (n = 57) and non-small cell lung cancer (n = 19). At first tumor evaluation 77 patients (46%) presented with immune unconfirmed progressive disease. Six patients (8%) experienced PSPD: 2 patients with partial response; 4 patients with stable disease. Increase in target lesions in the first CT-scan was more frequently associated to PSPD (67% vs 33%; P = .04). Patients with a PSPD had a superior survival when compared to patients progressing (median OS: 10.7 months vs 8.7 months; P = .07). Conclusions: A small subset of PSPD patients may experience response after an initial progression. Assessment of the current strategy for immune-related response evaluations may require further attention

    Ceralasertib (AZD6738), an oral ATR kinase inhibitor, in combination with carboplatin in patients with advanced solid tumors: a Phase I study

    No full text
    PURPOSE: This study reports the safety, tolerability, MTD, recommended phase II dose (RP2D), pharmacokinetic/pharmacodynamic profile, and preliminary antitumor activity of ceralasertib combined with carboplatin in patients with advanced solid tumors. It also examined exploratory predictive and pharmacodynamic biomarkers. PATIENTS AND METHODS: Eligible patients (n = 36) received a fixed dose of carboplatin (AUC5) with escalating doses of ceralasertib (20 mg twice daily to 60 mg once daily) in 21-day cycles. Sequential and concurrent combination dosing schedules were assessed. RESULTS: Two ceralasertib MTD dose schedules, 20 mg twice daily on days 4–13 and 40 mg once daily on days 1–2, were tolerated with carboplatin AUC5; the latter was declared the RP2D. The most common treatment-emergent adverse events (Common Terminology Criteria for Adverse Events grade ≄3) were anemia (39%), thrombocytopenia (36%), and neutropenia (25%). Dose-limiting toxicities of grade 4 thrombocytopenia (n = 2; including one grade 4 platelet count decreased) and a combination of grade 4 thrombocytopenia and grade 3 neutropenia occurred in 3 patients. Ceralasertib was quickly absorbed (t(max) ∌1 hour), with a terminal plasma half-life of 8–11 hours. Upregulation of pRAD50, indicative of ataxia telangiectasia mutated (ATM) activation, was observed in tumor biopsies during ceralasertib treatment. Two patients with absent or low ATM or SLFN11 protein expression achieved confirmed RECIST v1.1 partial responses. Eighteen of 34 (53%) response-evaluable patients had RECIST v1.1 stable disease. CONCLUSIONS: The RP2D for ceralasertib plus carboplatin was established as ceralasertib 40 mg once daily on days 1–2 administered with carboplatin AUC5 every 3 weeks, with pharmacokinetic and pharmacodynamic studies confirming pharmacodynamic modulation and preliminary evidence of antitumor activity observed

    La déficience de PBRM1 confÚre une létalité synthétique aux inhibiteurs de la réparation de l'ADN dans le cancer

    No full text
    International audienceInactivation of Polybromo 1 (PBRM1), a specific subunit of the PBAF chromatin remodeling complex, occurs frequently in cancer, including 40% of clear cell renal cell carcinomas (ccRCC). To identify novel therapeutic approaches to targeting PBRM1-defective cancers, we used a series of orthogonal functional genomic screens that identified PARP and ATR inhibitors as being synthetic lethal with PBRM1 deficiency. The PBRM1/PARP inhibitor synthetic lethality was recapitulated using several clinical PARP inhibitors in a series of in vitro model systems and in vivo in a xenograft model of ccRCC. In the absence of exogenous DNA damage, PBRM1-defective cells exhibited elevated levels of replication stress, micronuclei, and R-loops. PARP inhibitor exposure exacerbated these phenotypes. Quantitative mass spectrometry revealed that multiple R-loop processing factors were downregulated in PBRM1-defective tumor cells. Exogenous expression of the R-loop resolution enzyme RNase H1 reversed the sensitivity of PBRM1-deficient cells to PARP inhibitors, suggesting that excessive levels of R-loops could be a cause of this synthetic lethality. PARP and ATR inhibitors also induced cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS/STING) innate immune signaling in PBRM1-defective tumor cells. Overall, these findings provide the preclinical basis for using PARP inhibitors in PBRM1-defective cancers. SIGNIFICANCE: This study demonstrates that PARP and ATR inhibitors are synthetic lethal with the loss of PBRM1, a PBAF-specific subunit, thus providing the rationale for assessing these inhibitors in patients with PBRM1-defective cancer.L'inactivation de Polybromo 1 (PBRM1), une sous-unitĂ© spĂ©cifique du complexe de remodelage de la chromatine PBAF, se produit frĂ©quemment dans le cancer, y compris dans 40% des carcinomes rĂ©naux Ă  cellules claires (ccRCC). Afin d'identifier de nouvelles approches thĂ©rapeutiques pour cibler les cancers dĂ©ficients en PBRM1, nous avons utilisĂ© une sĂ©rie de cribles gĂ©nomiques fonctionnels orthogonaux qui ont identifiĂ© les inhibiteurs PARP et ATR comme Ă©tant synthĂ©tiquement lĂ©taux en cas de dĂ©ficience en PBRM1. La lĂ©talitĂ© synthĂ©tique des inhibiteurs de PBRM1/PARP a Ă©tĂ© rĂ©capitulĂ©e en utilisant plusieurs inhibiteurs cliniques de PARP dans une sĂ©rie de systĂšmes modĂšles in vitro et in vivo dans un modĂšle de xĂ©nogreffe de ccRCC. En l'absence de lĂ©sions exogĂšnes de l'ADN, les cellules dĂ©ficientes en PBRM1 prĂ©sentaient des niveaux Ă©levĂ©s de stress de rĂ©plication, de micronoyaux et de boucles R. L'exposition Ă  un inhibiteur de PARP a exacerbĂ© la lĂ©talitĂ© synthĂ©tique. L'exposition Ă  un inhibiteur de PARP a exacerbĂ© ces phĂ©notypes. La spectromĂ©trie de masse quantitative a rĂ©vĂ©lĂ© que plusieurs facteurs de traitement des boucles R Ă©taient rĂ©gulĂ©s Ă  la baisse dans les cellules tumorales dĂ©fectueuses de PBRM1. L'expression exogĂšne de l'enzyme de rĂ©solution des boucles R, la RNase H1, a inversĂ© la sensibilitĂ© des cellules PBRM1 dĂ©ficientes aux inhibiteurs de la PARP, ce qui suggĂšre que des niveaux excessifs de boucles R pourraient ĂȘtre une cause de cette lĂ©talitĂ© synthĂ©tique. Les inhibiteurs de PARP et d'ATR ont Ă©galement induit une signalisation immunitaire innĂ©e de type GMP cyclique-AMP synthase/stimulateur des gĂšnes de l'interfĂ©ron (cGAS/STING) dans les cellules tumorales dĂ©ficientes en PBRM1. Dans l'ensemble, ces rĂ©sultats fournissent une base prĂ©clinique pour l'utilisation des inhibiteurs de PARP dans les cancers dĂ©ficients en PBRM1. SIGNIFICATION : Cette Ă©tude dĂ©montre que les inhibiteurs de PARP et d'ATR sont synthĂ©tiquement lĂ©taux en cas de perte de PBRM1, une sous-unitĂ© spĂ©cifique du PBAF, ce qui justifie l'Ă©valuation de ces inhibiteurs chez les patients atteints d'un cancer dĂ©ficient en PBRM1
    corecore