61 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

    Chemosensitivity profiling of osteosarcoma tumour cell lines identifies a model of BRCAness

    Get PDF
    Osteosarcoma (OS) is an aggressive sarcoma, where novel treatment approaches are required. Genomic studies suggest that a subset of OS, including OS tumour cell lines (TCLs), exhibit genomic loss of heterozygosity (LOH) patterns reminiscent of BRCA1 or BRCA2 mutant tumours. This raises the possibility that PARP inhibitors (PARPi), used to treat BRCA1/2 mutant cancers, could be used to target OS. Using high-throughput drug sensitivity screening we generated chemosensitivity profiles for 79 small molecule inhibitors, including three clinical PARPi. Drug screening was performed in 88 tumour cell lines, including 18 OS TCLs. This identified known sensitivity effects in OS TCLs, such as sensitivity to FGFR inhibitors. When compared to BRCA1/2 mutant TCLs, OS TCLs, with the exception of LM7, were PARPi resistant, including those with previously determined BRCAness LoH profiles. Post-screen validation experiments confirmed PARPi sensitivity in LM7 cells as well as a defect in the ability to form nuclear RAD51 foci in response to DNA damage. LM7 provides one OS model for the study of PARPi sensitivity through a potential defect in RAD51-mediated DNA repair. The drug sensitivity dataset we generated in 88 TCLs could also serve as a resource for the study of drug sensitivity effects in OS

    Etude de la déficience en ERCC1 dans le cancer bronchique non-à-petites cellules et recherche de léthalité synthétique

    No full text
    Excision Repair Cross-Complementation group 1 (ERCC1) is a DNA repair enzyme that is frequently deficient in non-small cell lung cancer (NSCLC). Although low ERCC1 expression correlates with platinum sensitivity, the clinical effectiveness of platinum therapy is limited - mainly by toxicities and occurrence of resistance - highlighting the need for alternative treatment strategies. In addition, the lack of a reliable assay evaluating ERCC1 functionality in the clinical setting currently precludes personalising therapy based on ERCC1 status. To discover new synthetic lethality-based therapeutic strategies for ERCC1-defective tumours, high-throughput drug and siRNA screens in an isogenic NSCLC model of ERCC1 deficiency were performed. This approach identified multiple clinical poly(ADP-ribose) polymerase 1 and 2 (PARP1/2) inhibitors such as olaparib (AZD-2281), niraparib (MK-4827) and BMN 673 as being selective for ERCC1 deficiency. The mechanism underlying ERCC1-selective effects was dissected by studying molecular biomarkers of tumour cell response, and revealed that: (i) ERCC1-deficient cells displayed a significant delay in double-strand break repair associated with a profound and prolonged G2/M arrest following PARP1/2 inhibitor treatment; (ii) ERCC1 isoform 202, which has recently been shown to mediate platinum sensitivity, also modulated PARP1/2 sensitivity; (iii) ERCC1-deficiency was epistatic with homologous recombination deficiency, although ERCC1-deficient cells did not display a defect in RAD51 foci formation. This suggests that ERCC1 might be required to process PARP1/2 inhibitor induced DNA lesions prior to DNA strand invasion; and (iv) PARP1 silencing restored PARP1/2 inhibitor resistance in ERCC1-deficient cells but had no effect in ERCC1-proficient cells, supporting the hypothesis that PARP1 might be required for the ERCC1 selectivity of PARP1/2 inhibitors. This study indicated that PARP1/2 inhibitors as a monotherapy could represent a novel therapeutic strategy for NSCLC patients with ERCC1-deficient tumours, and a clinical protocol is being written to evaluate this hypothesis.To investigate whether a surrogate biomarker of ERCC1 functionality could be developed, four parallel approaches were undertaken in the ERCC1-isogenic NSCLC model: (i) UV irradiation, to evaluate the Nucleotide Excision Repair (NER) pathway; (ii) whole exome sequencing, to look for an ERCC1-associated genomic scar at the DNA level; (iii) transcriptomic analysis, to investigate changes at the RNA expression level; and (iv) SILAC (Stable Isotope Labeling by Amino acids in Cell culture) analysis, to compare proteomic profiles between ERCC1-proficient and ERCC1-deficient cells. These approaches allowed the identification of putative genomic signature and potential metabolic surrogate biomarkers - guanine deaminase (GDA) and nicotinamide phosphoribosyltransferase (NAMPT). Further validation and mechanistic investigations of these latter preliminary observations are warranted.Excision Repair Cross-Complementation group 1 (ERCC1) est une enzyme de rĂ©paration de l’ADN frĂ©quemment dĂ©ficiente dans le cancer bronchique non-Ă -petites cellules. Bien qu’une expression faible d’ERCC1 soit prĂ©dictive de rĂ©ponse aux sels de platine, l’efficacitĂ© des chimiothĂ©rapies Ă  base de platine est limitĂ©e par leur toxicitĂ© et l’apparition de rĂ©sistance, justifiant la nĂ©cessitĂ© de stratĂ©gies thĂ©rapeutiques alternatives. Par ailleurs, l’absence de test compagnon diagnostic permettant d’évaluer la fonctionnalitĂ© d’ERCC1 dans la pratique clinique empĂȘche actuellement toute thĂ©rapie personnalisĂ©e basĂ©e sur le statut ERCC1.Afin d’identifier de nouvelles stratĂ©gies thĂ©rapeutiques pour les tumeurs ERCC1-dĂ©ficientes en exploitant le concept de lĂ©talitĂ© synthĂ©tique, des screens Ă  haut-dĂ©bit , utilisant des composĂ©s pharmaceutiques ou par ARN interfĂ©rence, ont Ă©tĂ© rĂ©alisĂ©s dans un modĂšle isogĂ©nique de CBNPC dĂ©ficient en ERCC1. Cette approche a permis d’identifier plusieurs inhibiteurs de poly(ADP-ribose) polymerase 1 et 2 (PARP1/2), tels l’opalarib (AZD2281), le niraparib (MK-24827) et BMN 673 comme sĂ©lectifs pour les cellules ERCC1-dĂ©ficientes. Les mĂ©canismes sous-tendant cette sensibilitĂ© sĂ©lective ont Ă©tĂ© Ă©tudiĂ©s, et les rĂ©sultats suivants ont Ă©tĂ© mis en Ă©vidence : (i) les cellules ERCC1-dĂ©ficientes prĂ©sentent un blocage prolongĂ© en phase G2/M aprĂšs exposition Ă  l’olaparib ; (ii) l’isoforme 202 d’ERCC1, dont le rĂŽle a Ă©tĂ© rĂ©cemment mis en Ă©vidence dans la rĂ©sistance aux sels de platine, module Ă©galement la sensibilitĂ© aux inhibiteurs de PARP ; (iii) la dĂ©ficience en ERCC1 est Ă©pistatique avec les dĂ©fauts de recombinaison homologue (RH), malgrĂ© une capacitĂ© normale des cellules ERCC1-dĂ©ficientes Ă  former des foyers RAD51 ; ceci suggĂšre qu’ERCC1 pourrait intervenir dans la rĂ©paration d’une lĂ©sion de l’ADN induite par l’inhibiteur de PARP1/2 en amont de l’invasion du brin d’ADN lors de la RH ; (iv) l’inhibition de l’expression de PARP1 par ARN interfĂ©rence permet de restaurer la rĂ©sistance aux inhibiteurs de PARP1/2, dans les cellules ERCC1-dĂ©ficientes uniquement. Ces rĂ©sultats suggĂšrent que les inhibiteurs de PARP1/2 pourraient reprĂ©senter une nouvelle stratĂ©gie thĂ©rapeutique chez les patients dont la tumeur est dĂ©ficiente en ERCC1 et un essai clinique va ĂȘtre mis en place pour Ă©valuer cette hypothĂšse.Afin d’explorer la prĂ©sence de biomarqueurs de la fonctionnalitĂ© d’ERCC1, quatre approches ont Ă©tĂ© entreprises en parallĂšle dans le modĂšle isogĂ©nique de CBNPC dĂ©ficient en ERCC1: (i) irradiation aux UV, afin d’évaluer la voie NER (Nucleotide Excision Repair); (ii) sĂ©quençage d’exome, dans le but de rechercher une signature gĂ©nomique (ADN) ; (iii) analyse du transcriptome cellulaire, pour identifier des modifications d’expression d’ARN ; et (iv) SILAC (Stable Isotope Labeling by Amino acids in Cell culture) afin de comparer le protĂ©ome des cellules ERCC1-dĂ©ficientes et ERCC1-proficientes. Ces approches ont permis d’identifier une potentielle signature gĂ©nomique, ainsi que de biomarqueurs d’activitĂ© – guanine deaminase (GDA) et nicotinamide phosphoribosyltransferase (NAMPT). De plus amples validations et investigations mĂ©canistiques de ces observations prĂ©liminaires sont actuellement requises

    Etude de la déficience en ERCC1 dans le cancer bronchique non-à-petites cellules et recherche de léthalité synthétique

    No full text
    Excision Repair Cross-Complementation group 1 (ERCC1) est une enzyme de rĂ©paration de l ADN frĂ©quemment dĂ©ficiente dans le cancer bronchique non-Ă -petites cellules. Bien qu une expression faible d ERCC1 soit prĂ©dictive de rĂ©ponse aux sels de platine, l efficacitĂ© des chimiothĂ©rapies Ă  base de platine est limitĂ©e par leur toxicitĂ© et l apparition de rĂ©sistance, justifiant la nĂ©cessitĂ© de stratĂ©gies thĂ©rapeutiques alternatives. Par ailleurs, l absence de test compagnon diagnostic permettant d Ă©valuer la fonctionnalitĂ© d ERCC1 dans la pratique clinique empĂȘche actuellement toute thĂ©rapie personnalisĂ©e basĂ©e sur le statut ERCC1.Afin d identifier de nouvelles stratĂ©gies thĂ©rapeutiques pour les tumeurs ERCC1-dĂ©ficientes en exploitant le concept de lĂ©talitĂ© synthĂ©tique, des screens Ă  haut-dĂ©bit , utilisant des composĂ©s pharmaceutiques ou par ARN interfĂ©rence, ont Ă©tĂ© rĂ©alisĂ©s dans un modĂšle isogĂ©nique de CBNPC dĂ©ficient en ERCC1. Cette approche a permis d identifier plusieurs inhibiteurs de poly(ADP-ribose) polymerase 1 et 2 (PARP1/2), tels l opalarib (AZD2281), le niraparib (MK-24827) et BMN 673 comme sĂ©lectifs pour les cellules ERCC1-dĂ©ficientes. Les mĂ©canismes sous-tendant cette sensibilitĂ© sĂ©lective ont Ă©tĂ© Ă©tudiĂ©s, et les rĂ©sultats suivants ont Ă©tĂ© mis en Ă©vidence : (i) les cellules ERCC1-dĂ©ficientes prĂ©sentent un blocage prolongĂ© en phase G2/M aprĂšs exposition Ă  l olaparib ; (ii) l isoforme 202 d ERCC1, dont le rĂŽle a Ă©tĂ© rĂ©cemment mis en Ă©vidence dans la rĂ©sistance aux sels de platine, module Ă©galement la sensibilitĂ© aux inhibiteurs de PARP ; (iii) la dĂ©ficience en ERCC1 est Ă©pistatique avec les dĂ©fauts de recombinaison homologue (RH), malgrĂ© une capacitĂ© normale des cellules ERCC1-dĂ©ficientes Ă  former des foyers RAD51 ; ceci suggĂšre qu ERCC1 pourrait intervenir dans la rĂ©paration d une lĂ©sion de l ADN induite par l inhibiteur de PARP1/2 en amont de l invasion du brin d ADN lors de la RH ; (iv) l inhibition de l expression de PARP1 par ARN interfĂ©rence permet de restaurer la rĂ©sistance aux inhibiteurs de PARP1/2, dans les cellules ERCC1-dĂ©ficientes uniquement. Ces rĂ©sultats suggĂšrent que les inhibiteurs de PARP1/2 pourraient reprĂ©senter une nouvelle stratĂ©gie thĂ©rapeutique chez les patients dont la tumeur est dĂ©ficiente en ERCC1 et un essai clinique va ĂȘtre mis en place pour Ă©valuer cette hypothĂšse.Afin d explorer la prĂ©sence de biomarqueurs de la fonctionnalitĂ© d ERCC1, quatre approches ont Ă©tĂ© entreprises en parallĂšle dans le modĂšle isogĂ©nique de CBNPC dĂ©ficient en ERCC1: (i) irradiation aux UV, afin d Ă©valuer la voie NER (Nucleotide Excision Repair); (ii) sĂ©quençage d exome, dans le but de rechercher une signature gĂ©nomique (ADN) ; (iii) analyse du transcriptome cellulaire, pour identifier des modifications d expression d ARN ; et (iv) SILAC (Stable Isotope Labeling by Amino acids in Cell culture) afin de comparer le protĂ©ome des cellules ERCC1-dĂ©ficientes et ERCC1-proficientes. Ces approches ont permis d identifier une potentielle signature gĂ©nomique, ainsi que de biomarqueurs d activitĂ© guanine deaminase (GDA) et nicotinamide phosphoribosyltransferase (NAMPT). De plus amples validations et investigations mĂ©canistiques de ces observations prĂ©liminaires sont actuellement requises.Excision Repair Cross-Complementation group 1 (ERCC1) is a DNA repair enzyme that is frequently deficient in non-small cell lung cancer (NSCLC). Although low ERCC1 expression correlates with platinum sensitivity, the clinical effectiveness of platinum therapy is limited - mainly by toxicities and occurrence of resistance - highlighting the need for alternative treatment strategies. In addition, the lack of a reliable assay evaluating ERCC1 functionality in the clinical setting currently precludes personalising therapy based on ERCC1 status. To discover new synthetic lethality-based therapeutic strategies for ERCC1-defective tumours, high-throughput drug and siRNA screens in an isogenic NSCLC model of ERCC1 deficiency were performed. This approach identified multiple clinical poly(ADP-ribose) polymerase 1 and 2 (PARP1/2) inhibitors such as olaparib (AZD-2281), niraparib (MK-4827) and BMN 673 as being selective for ERCC1 deficiency. The mechanism underlying ERCC1-selective effects was dissected by studying molecular biomarkers of tumour cell response, and revealed that: (i) ERCC1-deficient cells displayed a significant delay in double-strand break repair associated with a profound and prolonged G2/M arrest following PARP1/2 inhibitor treatment; (ii) ERCC1 isoform 202, which has recently been shown to mediate platinum sensitivity, also modulated PARP1/2 sensitivity; (iii) ERCC1-deficiency was epistatic with homologous recombination deficiency, although ERCC1-deficient cells did not display a defect in RAD51 foci formation. This suggests that ERCC1 might be required to process PARP1/2 inhibitor induced DNA lesions prior to DNA strand invasion; and (iv) PARP1 silencing restored PARP1/2 inhibitor resistance in ERCC1-deficient cells but had no effect in ERCC1-proficient cells, supporting the hypothesis that PARP1 might be required for the ERCC1 selectivity of PARP1/2 inhibitors. This study indicated that PARP1/2 inhibitors as a monotherapy could represent a novel therapeutic strategy for NSCLC patients with ERCC1-deficient tumours, and a clinical protocol is being written to evaluate this hypothesis.To investigate whether a surrogate biomarker of ERCC1 functionality could be developed, four parallel approaches were undertaken in the ERCC1-isogenic NSCLC model: (i) UV irradiation, to evaluate the Nucleotide Excision Repair (NER) pathway; (ii) whole exome sequencing, to look for an ERCC1-associated genomic scar at the DNA level; (iii) transcriptomic analysis, to investigate changes at the RNA expression level; and (iv) SILAC (Stable Isotope Labeling by Amino acids in Cell culture) analysis, to compare proteomic profiles between ERCC1-proficient and ERCC1-deficient cells. These approaches allowed the identification of putative genomic signature and potential metabolic surrogate biomarkers - guanine deaminase (GDA) and nicotinamide phosphoribosyltransferase (NAMPT). Further validation and mechanistic investigations of these latter preliminary observations are warranted.PARIS11-SCD-Bib. Ă©lectronique (914719901) / SudocSudocFranceF

    Immunotherapy for SMARCB1-Deficient Sarcomas: Current Evidence and Future Developments

    No full text
    Mutations in subunits of the SWItch Sucrose Non-Fermentable (SWI/SNF) complex occur in 20% of all human tumors. Among these, the core subunit SMARCB1 is the most frequently mutated, and SMARCB1 loss represents a founder driver event in several malignancies, such as malignant rhabdoid tumors (MRT), epithelioid sarcoma, poorly differentiated chordoma, and renal medullary carcinoma (RMC). Intriguingly, SMARCB1-deficient pediatric MRT and RMC have recently been reported to be immunogenic, despite their very simple genome and low tumor mutational burden. Responses to immune checkpoint inhibitors have further been reported in some SMARCB1-deficient diseases. Here, we will review the preclinical data and clinical data that suggest that immunotherapy, including immune checkpoint inhibitors, may represent a promising therapeutic strategy for SMARCB1-defective tumors. We notably discuss the heterogeneity that exists among the spectrum of malignancies driven by SMARCB1-loss, and highlight challenges that are at stake for developing a personalized immunotherapy for these tumors, notably using molecular profiling of the tumor and of its microenvironment

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

    Get PDF
    Desenvolupament de fàrmacs; MelanomaDesarrollo de fármacos; MelanomaDrug development; MelanomaBackground 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.The study was sponsored by Orion Corporation, Orion Pharma, Espoo, Finland, the developer of ODM-207

    Targeting the DNA damage response in immuno-oncology: developments and opportunities

    No full text
    International audienceImmunotherapy has revolutionized cancer treatment and substantially improved patient outcome with regard to multiple tumour types. However, most patients still do not benefit from such therapies, notably because of the absence of pre-existing T cell infiltration. DNA damage response (DDR) deficiency has recently emerged as an important determinant of tumour immunogenicity. A growing body of evidence now supports the concept that DDR-targeted therapies can increase the antitumour immune response by (1) promoting antigenicity through increased mutability and genomic instability, (2) enhancing adjuvanticity through the activation of cytosolic immunity and immunogenic cell death and (3) favouring reactogenicity through the modulation of factors that control the tumour–immune cell synapse. In this Review, we discuss the interplay between the DDR and anticancer immunity and highlight how this dynamic interaction contributes to shaping tumour immunogenicity. We also review the most innovative preclinical approaches that could be used to investigate such effects, including recently developed ex vivo systems. Finally, we highlight the therapeutic opportunities presented by the exploitation of the DDR–anticancer immunity interplay, with a focus on those in early-phase clinical development
    • 

    corecore