Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.865
Filtrer
1.
J Immunother Cancer ; 12(10)2024 Oct 04.
Article de Anglais | MEDLINE | ID: mdl-39366751

RÉSUMÉ

BACKGROUND: In addition to their established action of synthetic lethality in tumor cells, poly(ADP-ribose) polymerase inhibitors (PARPis) also orchestrate tumor immune microenvironment (TIME) that contributes to suppressing tumor growth. However, it remains not fully understood whether and how PARPis trigger tumor-targeting immune responses. METHODS: To decode the immune responses reshaped by PARPis, we conducted T-cell receptor (TCR) sequencing and immunohistochemical (IHC) analyses of paired clinical specimens before and after niraparib monotherapy obtained from a prospective study, as well as ID8 mouse ovarian tumors. To validate the induction of immunogenic cell death (ICD) by PARPis, we performed immunofluorescence/IHC staining with homologous recombination deficiency tumor cells and patient-derived xenograft tumor tissues, respectively. To substantiate that PARPis elicited tumor cell pyroptosis, we undertook comprehensive assessments of the cellular morphological features, cleavage of gasdermin (GSDM) proteins, and activation of TNF-caspase signaling pathways through genetic downregulation/depletion and selective inhibition. We also evaluated the critical role of pyroptosis in tumor suppression and immune activation following niraparib treatment using a syngeneic mouse model with implanting CRISPR/Cas9 edited Gsdme-/ - ID8 tumor cells into C57BL/6 mice. RESULTS: Our findings revealed that PARPis augmented the proportion of neoantigen-recognized TCR clones and TCR clonal expansion, and induced an inflamed TIME characterized by increased infiltration of both innate and adaptive immune cells. This PARPis-strengthened immune response was associated with the induction of ICD, specifically identified as pyroptosis, which possessed distinctive morphological features and GSDMD/E cleavage. It was validated that the cleavage of GSDMD/E was due to elevated caspase 8 activity downstream of the TNFR1, rather than FAS and TRAIL-R. On PARP inhibition, the NF-κB signaling pathway was activated, leading to increased secretion of TNF-α and subsequent initiation of the TNFR1-caspase 8 cascade. Impeding pyroptosis through the depletion of Gsdme significantly compromised the tumor-suppressing effects of PARP inhibition and undermined the anti-immune response in the syngeneic ID8 mouse model. CONCLUSIONS: PARPis induce a specific type of ICD called pyroptosis via TNF-caspase 8-GSDMD/E axis, resulting in an inflamed TIME and augmentation of tumor-targeting immune responses. These findings deepen our understanding of PARPis activities and point toward a promising avenue for synergizing PARPis with immunotherapeutic interventions. TRIAL REGISTRATION NUMBER: NCT04507841.


Sujet(s)
Tumeurs de l'ovaire , Inhibiteurs de poly(ADP-ribose) polymérases , Pyroptose , Femelle , Pyroptose/effets des médicaments et des substances chimiques , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Animaux , Humains , Souris , Protéines de liaison aux phosphates/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Transduction du signal , Lignée cellulaire tumorale , Microenvironnement tumoral , Pipéridines/pharmacologie , Pipéridines/usage thérapeutique , Gasdermines
2.
ESMO Open ; 9(9): 103684, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39255537

RÉSUMÉ

BACKGROUND: Controlled trials have consistently demonstrated the efficacy of poly(ADP-ribose) polymerase inhibitors (PARPis) in patients with metastatic castration-resistant prostate cancer (mCRPC) and BRCA1 or BRCA2 alterations (BRCAalt). However, the reported efficacy of PARPi for alterations in other homologous recombination repair (HRR) genes is less consistent. We sought to evaluate the routine practice effectiveness of PARPi between and within these groups. DESIGN: Patient-level data from a deidentified nationwide (USA-based) cancer clinico-genomic database between January 2011 and September 2023 were extracted. Patients with mCRPC and comprehensive genomic profiling by liquid biopsy [circulating tumor DNA (ctDNA)] or tissue (tumor) biopsy and who received single-agent PARPi were included and grouped by BRCAalt, ATMalt, other HRR, or no HRR. We further subcategorized BRCAalt into homozygous loss (BRCAloss) and all other deleterious BRCAalt (otherBRCAalt). RESULTS: A total of 445 patients met inclusion criteria: 214 with tumor and 231 with ctDNA. BRCAalt had more favorable outcomes to PARPi compared with ATM, other HRR, and no HRR groups. Within the BRCAalt subgroup, compared with other BRCAalt, BRCAloss had a more favorable time to next treatment (median 9 versus 19.4 months, P = 0.005), time to treatment discontinuation (median 8 versus 14 months, P = 0.006), and routine practice overall survival (median 14.7 versus 19.4 months, P = 0.016). Tumor BRCAloss prevalence (3.1%) was similar to ctDNA prevalence in liquid biopsy specimens with high tumor fraction (>20%). BRCAloss was not detected in orthogonal germline testing. CONCLUSIONS: PARPi routine practice effectiveness between groups mirrors prospective trials. Within the BRCAalt group, BRCAloss had the best outcomes. Unless the ctDNA tumor fraction is very high, somatic tissue testing (archival or metastatic) should be prioritized to identify patients who may benefit most from PARPi. When tissue testing is not clinically feasible, sufficient ctDNA tumor fraction levels for detection are enriched at clinical timepoints associated with tumor progression.


Sujet(s)
Séquençage nucléotidique à haut débit , Inhibiteurs de poly(ADP-ribose) polymérases , Tumeurs prostatiques résistantes à la castration , Humains , Mâle , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Sujet âgé , Séquençage nucléotidique à haut débit/méthodes , Marqueurs biologiques tumoraux/génétique , Protéine BRCA2/génétique , Adulte d'âge moyen , ADN tumoral circulant/génétique , Biopsie liquide/méthodes , Protéine BRCA1/génétique , Métastase tumorale
3.
Ann Med ; 56(1): 2399759, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39258876

RÉSUMÉ

BACKGROUND: The status of BRCA1/2 genes plays a crucial role in the treatment decision-making process for multiple cancer types. However, due to high costs and limited resources, a demand for BRCA1/2 genetic testing among patients is currently unmet. Notably, not all patients with BRCA1/2 mutations achieve favorable outcomes with poly (ADP-ribose) polymerase inhibitors (PARPi), indicating the necessity for risk stratification. In this study, we aimed to develop and validate a multimodal model for predicting BRCA1/2 gene status and prognosis with PARPi treatment. METHODS: We included 1695 slides from 1417 patients with ovarian, breast, prostate, and pancreatic cancers across three independent cohorts. Using a self-attention mechanism, we constructed a multi-instance attention model (MIAM) to detect BRCA1/2 gene status from hematoxylin and eosin (H&E) pathological images. We further combined tissue features from the MIAM model, cell features, and clinical factors (the MIAM-C model) to predict BRCA1/2 mutations and progression-free survival (PFS) with PARPi therapy. Model performance was evaluated using area under the curve (AUC) and Kaplan-Meier analysis. Morphological features contributing to MIAM-C were analyzed for interpretability. RESULTS: Across the four cancer types, MIAM-C outperformed the deep learning-based MIAM in identifying the BRCA1/2 genotype. Interpretability analysis revealed that high-attention regions included high-grade tumors and lymphocytic infiltration, which correlated with BRCA1/2 mutations. Notably, high lymphocyte ratios appeared characteristic of BRCA1/2 mutations. Furthermore, MIAM-C predicted PARPi therapy response (log-rank p < 0.05) and served as an independent prognostic factor for patients with BRCA1/2-mutant ovarian cancer (p < 0.05, hazard ratio:0.4, 95% confidence interval: 0.16-0.99). CONCLUSIONS: The MIAM-C model accurately detected BRCA1/2 gene status and effectively stratified prognosis for patients with BRCA1/2 mutations.


Sujet(s)
Mutation , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Femelle , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Mâle , Protéine BRCA1/génétique , Protéine BRCA2/génétique , Pronostic , Adulte d'âge moyen , Survie sans progression , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Thérapie moléculaire ciblée/méthodes , Tumeurs du pancréas/génétique , Tumeurs du pancréas/traitement médicamenteux , Adulte
4.
Pharmacoepidemiol Drug Saf ; 33(9): e70001, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39252433

RÉSUMÉ

PURPOSE: This retrospective real-world study compared overall survival (OS) between patients with BRCA wild-type (BRCAwt) recurrent epithelial ovarian cancer (OC) who received niraparib second-line maintenance (2LM) versus active surveillance (AS) using target trial emulation, cloning, inverse probability of censoring weighting (IPCW) methodology to minimize immortal time bias. METHODS: Eligible patients from a United States-based, deidentified, electronic health record-derived database were diagnosed with epithelial OC (January 1, 2011-May 31, 2021), were BRCAwt, and completed second-line (2L) therapy (January 1, 2017-March 2, 2022). Patient data were cloned at index (2L last treatment date), assigned to niraparib 2LM and AS cohorts, and censored when treatment deviated from clone assignment. Follow-up was measured from index to earliest of study end (May 31, 2022), last activity, or death. Median OS (mOS) and hazard ratios were estimated from stabilized IPCW Kaplan-Meier curves and Cox regression models. RESULTS: Overall, 199 patients received niraparib 2LM, and 707 had their care managed with AS. Key characteristics were balanced across cohorts after cloning and stabilized IPCW. Median follow-up was 15.6- and 9.3-months pre-cloning. IPCW mOS was 24.1 months (95% CI: 20.9-29.5) and 18.4 months (95% CI: 15.1-22.8) in niraparib 2LM and AS cohorts, respectively (hazard ratio, 0.77; 95% CI: 0.66-0.89). CONCLUSIONS: This real-world study provides supportive evidence of an OS benefit for patients with BRCAwt recurrent OC who received 2LM niraparib monotherapy compared with those whose care was managed with AS. The analytic strategies implemented were useful in minimizing immortal time bias and measured confounding.


Sujet(s)
Indazoles , Récidive tumorale locale , Tumeurs de l'ovaire , Pipéridines , Humains , Femelle , Pipéridines/usage thérapeutique , Pipéridines/administration et posologie , Indazoles/usage thérapeutique , Indazoles/administration et posologie , Adulte d'âge moyen , Études rétrospectives , Sujet âgé , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/mortalité , Carcinome épithélial de l'ovaire/traitement médicamenteux , Carcinome épithélial de l'ovaire/mortalité , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/administration et posologie , Adulte , Observation (surveillance clinique) , États-Unis/épidémiologie , Chimiothérapie de maintenance/méthodes , Bases de données factuelles
5.
J Gynecol Oncol ; 35(5): e114, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39251349

RÉSUMÉ

OBJECTIVE: To evaluate the long-term efficacy and safety of niraparib in Japanese women with heavily pretreated ovarian cancer. METHODS: This was the follow-up analysis of a phase 2, multicenter, open-label, single-arm study in Japanese women with homologous recombination-deficient, platinum-sensitive, relapsed, high-grade serous epithelial ovarian, fallopian tube, or primary peritoneal cancer who had completed 3-4 lines of chemotherapy and were poly(ADP-ribose) polymerase inhibitor naïve. Participants received niraparib (starting dose, 300 mg) once daily in continuous 28-day cycles until objective disease progression, unacceptable toxicity, or consent withdrawal. The primary endpoint was confirmed objective response rate (ORR), as assessed using Response Evaluation Criteria in Solid Tumors version 1.1. Safety evaluations included treatment-emergent adverse events (TEAEs). RESULTS: 20 patients were enrolled in the study and included in both efficacy and safety analyses. Median total study duration was 759.5 days. Median dose intensity was 201.3 mg/day. Confirmed ORR was 60.0% (90% confidence interval [CI]=39.4-78.3); 2 patients had complete response and 10 patients had partial response. Median duration of response was 9.9 months (95% CI=3.9-26.9) and the disease control rate was 90.0% (95% CI=68.3-98.8). The most common TEAEs were anemia (n=15), nausea (n=12), and decreased platelet count (n=11). TEAEs leading to study drug dose reduction, interruption, or discontinuation were reported in 16 (80.0%), 15 (75.0%), and 2 patients (10.0%), respectively. CONCLUSION: The long-term efficacy and safety profile of niraparib was consistent with previous findings in the equivalent population in non-Japanese patients. No new safety signals were identified. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT03759600.


Sujet(s)
Indazoles , Tumeurs de l'ovaire , Pipéridines , Inhibiteurs de poly(ADP-ribose) polymérases , Adulte , Sujet âgé , Femelle , Humains , Adulte d'âge moyen , Carcinome épithélial de l'ovaire/traitement médicamenteux , Peuples d'Asie de l'Est , Tumeurs de la trompe de Fallope/traitement médicamenteux , Recombinaison homologue , Indazoles/effets indésirables , Indazoles/usage thérapeutique , Japon , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/génétique , Tumeurs du péritoine/traitement médicamenteux , Phtalazines/effets indésirables , Phtalazines/usage thérapeutique , Pipéridines/effets indésirables , Pipéridines/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/effets indésirables , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique
7.
Cell ; 187(18): 4829-4830, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39241744

RÉSUMÉ

Homologous-recombination deficiency in DNA repair characterizes a unique group of cancers that are vulnerable to PARP inhibitors and cytotoxic chemotherapy. In this issue of Cell, Luo et al., demonstrated that this genetic attribute in cancer cells may reprogram tumor immune microenvironment and show promise of targeting effector-Treg cells.


Sujet(s)
Inhibiteurs de poly(ADP-ribose) polymérases , Microenvironnement tumoral , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Humains , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Animaux
8.
BMC Pulm Med ; 24(1): 448, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39272066

RÉSUMÉ

BACKGROUND: PARP inhibitors (PARPi) are used in the treatment of ovarian, breast, pancreatic, and prostate cancers. Pneumonitis has been identified as a potential side effect, with a higher meta-analysis-assessed risk for olaparib versus other PARPi. Olaparib-induced interstitial lung disease (O-ILD) was first described within the Japanese population, with few information available for Caucasian patients. METHODS: We performed a retrospective study by pooling data from the French and Belgian pharmacovigilance databases from 2018 to 2022. Patients with O-ILD were included following a central review by: 1) pharmacologists using the French drug causality assessment method; 2) senior pneumologists or radiologists, using the Fleischner Society's recommendations. RESULTS: Five patients were identified and analysed. All were females, with ovarian or breast cancer. Median age at O-ILD diagnosis was 71 (38-72) years old, with no smoking history. Median delay between treatment initiation and symptom occurrence was 12 (6-33) weeks. Pneumonitis severity assessed using the Common Terminology Criteria for Adverse Events V5 was Grade 3 (n = 4) or 2 (n = 1). CT-scan review (n = 3) described hypersensitivity pneumonitis reaction as a common pattern. Bronchioalveolar lavage (n = 4) revealed lymphocytic alveolitis. Treatments relied on olaparib discontinuation (n = 5) and glucocorticoid intake (n = 4), with no fatal issue. Safe re-challenge with PARPi occurred in two patients. Forty additional O-ILD cases were identified in the WHO VigiBase database, including one fatal case. CONCLUSIONS: PARPi-ILD is a rare but potentially life-threatening disease, presenting as a hypersensitivity pneumonitis pattern within 3 months of PARPi initiation. Treatment primarily relies on medication discontinuation. Re-challenging with another PARPi could be considered. CLINICAL TRIAL NUMBER: CEPRO #2023-010.


Sujet(s)
Pneumopathies interstitielles , Pharmacovigilance , Phtalazines , Pipérazines , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Pneumopathies interstitielles/induit chimiquement , Pneumopathies interstitielles/imagerie diagnostique , Phtalazines/effets indésirables , Phtalazines/usage thérapeutique , Femelle , Pipérazines/effets indésirables , Pipérazines/usage thérapeutique , Études rétrospectives , Sujet âgé , Adulte d'âge moyen , Adulte , Inhibiteurs de poly(ADP-ribose) polymérases/effets indésirables , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Tomodensitométrie , Tumeurs de l'ovaire/traitement médicamenteux , France , Belgique
9.
ESMO Open ; 9(9): 103694, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39232440

RÉSUMÉ

BACKGROUND: Poly(ADP-ribose) polymerase inhibitors (PARPis) improved advanced ovarian cancer treatment. Most patients progress during or following PARPi exposure, however, with concerns about sensitivity of subsequent chemotherapy. PATIENTS AND METHODS: In this international cohort study, we evaluated the efficacy of a subsequent chemotherapy following PARPi exposure in high-grade ovarian carcinoma patients. Endpoints included progression-free survival (PFS), overall survival and a multivariable Cox model was built to identify factors influencing PFS. RESULTS: We included 291 patients from four international centers treated between January 2002 and December 2021. The median number of previous chemotherapy was 1 (1.0-7.0), the median duration of PARPi exposure was 6.5 months (0.2-54.3 months). PARPi was used in first line in 14.1% patients. Most progressions occurred under PARPi exposure (89.1%). A BRCA pathogenic variant was identified in 130 patients (44.7%), absent in 157 patients (54.0%), and undocumented in 4 patients (1.4%). Platinum-based CT (PBC) and non-PBC were administered as subsequent treatments in, respectively, 182 patients (62.5%) and 109 patients (37.5%). Multivariable analyses showed that platinum-free interval (PFI) >6 months [adjusted hazards ratio (HR), 0.52; 95% confidence interval (CI) 0.39-0.70] and type of initial surgery (adjusted HR, 1.41; 95% CI 1.07-1.87; interval or closing surgery versus primary surgery) were associated with PFS, independent of BRCA status or line of therapy (≥2 versus 1). In patients with a PFI >6 months, PBC was numerically associated with the best PFS (adjusted HR, 0.68; 95% CI 0.46-1.01). CONCLUSION: This is the largest real-world study assessing the efficacy of subsequent chemotherapy in patients progressing during PARPi exposure. The patients have poor outcomes. PBC is the best option in patients progressing on PARPi and eligible for PBC rechallenge (PFI >6 months).


Sujet(s)
Tumeurs de l'ovaire , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Femelle , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/mortalité , Tumeurs de l'ovaire/génétique , Adulte d'âge moyen , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Sujet âgé , Évolution de la maladie , Adulte , Survie sans progression , Études rétrospectives , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Études de cohortes , Sujet âgé de 80 ans ou plus
10.
Chin Clin Oncol ; 13(Suppl 1): AB067, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39295385

RÉSUMÉ

BACKGROUND: Glioblastoma (GBM) is the most malignant brain tumor and ranks among the most lethal of all human cancers, without improvements in survival over the last 30 years. Data obtained in our group suggest that PARP1, a well-known DNA-repairing protein, could also play a key role in the regulation of cell cycle through its interaction with the transcription factor E2F1. Therefore, considering that most oncogenic processes are associated with cell cycle deregulation, we hypothesized that disruption of PARP1-E2F1 interaction would provide a novel therapeutic approach to different types of cancer. METHODS: The identification of novel compounds disrupting PARP1-E2F1 interaction was carried out by combining in silico and in vitro screening, using a rational drug design. The virtual screen was performed using a molecular library of several million compounds at the selected target site, using AtomNet® (Atomwise, San Francisco, CA, USA), the first deep learning neural network for structure-based drug design and discovery. Since there is no complete structural information of the PARP1-E2F1 protein-protein interaction, a homologous structure of the BRCT domain of BRCA1 complex with the phospho-peptide (PDBID: 1T2V) was used to identify the potential binding interface of BRCT domain of PARP-1 (PDBID: 2COK) and the E2F1 protein. Top scoring compounds were clustered and filtered to obtain a final subset of 83 compounds that were incorporated to our in vitro screening, which included both transcriptional E2F1 activity and survival studies. Complete culture medium supplemented with the compounds selected in the in silico screening (10 µM) were added and incubated for 24 hours. E2F1 activity was observed by measuring luminescence. For the viability assay, the fluorescence reading was performed (excitation 544 nm and emission 590 nm). RESULTS: The in silico and in vitro screening resulted in 12 compounds that inhibited E2F1 transcriptional activity and significantly reduced cell number. The highest inhibition of both E2F1 transcriptional activity and cell growth was observed with compound 3797, which was selected for further studies. CONCLUSIONS: Both in silico and in vitro results indicate that inhibition of PARP1-E2F1 transcriptional activity may provide a new rationale for designing novel therapeutic approaches for the treatment of GBM.


Sujet(s)
Facteur de transcription E2F1 , Glioblastome , Poly (ADP-Ribose) polymerase-1 , Humains , Glioblastome/traitement médicamenteux , Glioblastome/génétique , Poly (ADP-Ribose) polymerase-1/métabolisme , Facteur de transcription E2F1/métabolisme , Développement de médicament/méthodes , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/génétique , Lignée cellulaire tumorale , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique
11.
Anticancer Res ; 44(10): 4203-4211, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39348956

RÉSUMÉ

BACKGROUND/AIM: The emergence of novel DNA damage repair (DDR) pathways in molecular-target therapy drugs (MTTD) has shown promising outcomes in treating patients with metastatic castration-resistant prostate cancer (mCRPC). About 25% of mCRPC patients have actionable deleterious aberrations in DDR genes, primarily in the homologous recombination (HR) pathway. However, the response rate in patients with BRCA1/2 or mutations in HRR-related genes is only 45%-55%, when exposed to poly ADP ribose polymerase (PARP) inhibitor-based therapy (PARPi). A frequent characteristic feature of prostate cancer (PC) is the occurrence of genomic rearrangement that affects the transmembrane protease serine 2 (TMPRSS2) and E26 transformation-specific (ETS)- transcription factor-related gene (ERG). MATERIALS AND METHODS: In this study, a total of 114 patients with mCRPC had their RNA and DNA sequenced using next-generation sequencing. RESULTS: Based on their genetic profile of deleterious gene alterations of BRCA1/2 or ATM, six patients were selected for PARPi. Patients with TMPRSS2:ERG gene fusion and homozygous alteration in ATM or BRCA2 (n=2) or heterozygous alterations (BRCA1 or BRCA2) and lack of TMPRSS2:ERG gene fusion (n=2) did not show clinical benefit from PARPi (treatment duration <16 weeks). In contrast, patients (n=2) without TMPRSS2:ERG gene fusion and homozygous deleterious alterations in ATM or BRCA2 all had clinical benefit from PARPi (treatment duration ≥16 weeks). CONCLUSION: The TMPRSS2:ERG transcript product might be used as a PARPi resistance biomarker.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Inhibiteurs de poly(ADP-ribose) polymérases , Tumeurs prostatiques résistantes à la castration , Humains , Mâle , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Protéines de fusion oncogènes/génétique , Sujet âgé , Serine endopeptidases/génétique , Adulte d'âge moyen , Protéine BRCA2/génétique , Protéine BRCA1/génétique
12.
Cells ; 13(16)2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39195238

RÉSUMÉ

Uveal melanoma (UM) is the most common primary intraocular tumor in adults, with no standardized treatment for advanced disease. Based on preliminary bioinformatical analyses DTYMK and PARP1 were selected as potential therapeutic targets. High levels of both proteins were detected in uveal melanoma cells and correlated with increased tumor growth and poor prognosis. In vitro tests on MP41 (BAP1 positive) and MP46 (BAP1 negative) cancer cell lines using inhibitors pamiparib (PARP1) and Ymu1 (DTYMK) demonstrated significant cytotoxic effects. Combined treatment had synergistic effects in MP41 and additive in MP46 cell lines, reducing cell proliferation and inhibiting the mTOR signaling pathway. Furthermore, the applied inhibitors in combination decreased cell motility and migration speed, especially for BAP1-negative cell lines. Our hypothesis of the double hit into tumoral DNA metabolism as a possible therapeutic option in uveal melanoma was confirmed since combined targeting of DTYMK and PARP1 affected all tested cytophysiological parameters with the highest efficiency. Our in vitro findings provide insights into novel therapeutic avenues for managing uveal melanoma, warranting further exploration in preclinical and clinical settings.


Sujet(s)
Prolifération cellulaire , Mélanome , Poly (ADP-Ribose) polymerase-1 , Tumeurs de l'uvée , Humains , Tumeurs de l'uvée/traitement médicamenteux , Tumeurs de l'uvée/anatomopathologie , Tumeurs de l'uvée/métabolisme , Mélanome/traitement médicamenteux , Mélanome/anatomopathologie , Mélanome/métabolisme , Lignée cellulaire tumorale , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique
13.
Expert Rev Mol Med ; 26: e17, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39189367

RÉSUMÉ

ADP-ribosyltransferases of the PARP family encompass a group of enzymes with variegated regulatory functions in cells, ranging from DNA damage repair to the control of cell-cycle progression and immune response. Over the years, this knowledge has led to the use of PARP1/2 inhibitors as mainstay pharmaceutical strategies for the treatment of ovarian, pancreatic, prostate and breast cancers, holding mutations in genes encoding for proteins involved in the DNA repair mechanisms (synthetic lethality). Meanwhile, the last decade has witnessed significant progress in comprehending cellular pathways regulated by mono-ADP-ribosylation, with a huge effort in the development of novel selective compounds to inhibit those PARPs endowed with mono-ADP-ribosylation activity. This review focuses on the progress achieved in the cancer field, delving into most recent findings regarding the role of a subset of enzymes - the interferon-stimulated PARPs - in cancer progression.


Sujet(s)
ADP-Ribosylation , Interférons , Tumeurs , Inhibiteurs de poly(ADP-ribose) polymérases , Poly(ADP-ribose) polymerases , Transduction du signal , Humains , Tumeurs/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Interférons/métabolisme , Poly(ADP-ribose) polymerases/métabolisme , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Animaux , Réparation de l'ADN
14.
ESMO Open ; 9(9): 103685, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39214046

RÉSUMÉ

BACKGROUND: Among patients with advanced high-grade ovarian carcinoma (aHGOC) treated with poly (ADP-ribose) polymerase (PARP) inhibitors (PARPis), the presence of a germline BRCA pathogenic variant (gBRCA-PV) may increase the risk of bone marrow mutagenesis resulting in postcytotoxic therapy myelodysplastic neoplasms (MDS-pCT) or acute myeloid leukemia (AML-pCT), as it is expressed in heterozygosity also by hematopoietic progenitors. We aimed to investigate the occurrence of post-PARPi MDSs/AMLs-pCTs according to gBRCA-PV status. PATIENTS AND METHODS: We conducted a retrospective single-center study to evaluate MDS/AML-pCT in patients with aHGOC and a known gBRCA-PV status receiving at least 8 weeks of maintenance PARPi, in any line of therapy, from February 2017 to December 2022. The endpoint was the proportion of patients who experienced MDSs-pCT and AMLs-pCT during and after treatment with PARPi, in gBRCA-PV carriers and non-carriers. RESULTS: A total of 166 patients were included: 95 (57%) had a gBRCA-PV and 72% received PARPi for recurrent disease. The number of lines of chemotherapies before and after PARPi, median overall survival, and median follow-up were comparable between gBRCA-PV carriers and non-carriers. After a median follow-up of 40.0 (95% confidence interval: 35.7-44.3) months, 10 (6%) patients were diagnosed with an MDS-pCT and 4 (2%) with an AML-pCT. A higher proportion of MDSs/AMLs-pCT (10% versus 2%; P = 0.16) and, in particular, of MDSs-pCT (9% versus 1%; P = 0.04) was observed among gBRCA-PV carriers. CONCLUSIONS: The presence of a gBRCA-PV is associated with a higher risk of MDS-pCT and possibly of myeloid neoplasms after PARPi in patients with aHGOC who received PARPi, especially in the setting of recurrent disease.


Sujet(s)
Protéine BRCA1 , Protéine BRCA2 , Mutation germinale , Tumeurs de l'ovaire , Inhibiteurs de poly(ADP-ribose) polymérases , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Femelle , Humains , Adulte d'âge moyen , Protéine BRCA1/génétique , Protéine BRCA2/génétique , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/traitement médicamenteux , Syndromes myélodysplasiques/traitement médicamenteux , Syndromes myélodysplasiques/génétique , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/anatomopathologie , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Études rétrospectives
15.
Genome Med ; 16(1): 107, 2024 Aug 26.
Article de Anglais | MEDLINE | ID: mdl-39187844

RÉSUMÉ

BACKGROUND: Poly (ADP-ribose) polymerase 1 and 2 (PARP1/2) inhibitors (PARPi) are targeted therapies approved for homologous recombination repair (HRR)-deficient breast, ovarian, pancreatic, and prostate cancers. Since inhibition of PARP1 is sufficient to cause synthetic lethality in tumors with homologous recombination deficiency (HRD), PARP1 selective inhibitors such as saruparib (AZD5305) are being developed. It is expected that selective PARP1 inhibition leads to a safer profile that facilitates its combination with other DNA damage repair inhibitors. Here, we aimed to characterize the antitumor activity of AZD5305 in patient-derived preclinical models compared to the first-generation PARP1/2 inhibitor olaparib and to identify mechanisms of resistance. METHODS: Thirteen previously characterized patient-derived tumor xenograft (PDX) models from breast, ovarian, and pancreatic cancer patients harboring germline pathogenic alterations in BRCA1, BRCA2, or PALB2 were used to evaluate the efficacy of AZD5305 alone or in combination with carboplatin or an ataxia telangiectasia and Rad3 related (ATR) inhibitor (ceralasertib) and compared it to the first-generation PARPi olaparib. We performed DNA and RNA sequencing as well as protein-based assays to identify mechanisms of acquired resistance to either PARPi. RESULTS: AZD5305 showed superior antitumor activity than the first-generation PARPi in terms of preclinical complete response rate (75% vs. 37%). The median preclinical progression-free survival was significantly longer in the AZD5305-treated group compared to the olaparib-treated group (> 386 days vs. 90 days). Mechanistically, AZD5305 induced more replication stress and genomic instability than the PARP1/2 inhibitor olaparib in PARPi-sensitive tumors. All tumors at progression with either PARPi (39/39) showed increase of HRR functionality by RAD51 foci formation. The most prevalent resistance mechanisms identified were the acquisition of reversion mutations in BRCA1/BRCA2 and the accumulation of hypomorphic BRCA1. AZD5305 did not sensitize PDXs with acquired resistance to olaparib but elicited profound and durable responses when combined with carboplatin or ceralasertib in 3/6 and 5/5 models, respectively. CONCLUSIONS: Collectively, these results show that the novel PARP1 selective inhibitor AZD5305 yields a potent antitumor response in PDX models with HRD and delays PARPi resistance alone or in combination with carboplatin or ceralasertib, which supports its use in the clinic as a new therapeutic option.


Sujet(s)
Protéine BRCA1 , Protéine BRCA2 , Inhibiteurs de poly(ADP-ribose) polymérases , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Animaux , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Souris , Protéine BRCA1/génétique , Protéine BRCA2/génétique , Femelle , Phtalazines/pharmacologie , Phtalazines/usage thérapeutique , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Pipérazines/pharmacologie , Pipérazines/usage thérapeutique , Indoles/usage thérapeutique , Indoles/pharmacologie , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Carboplatine/pharmacologie , Carboplatine/usage thérapeutique , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique
16.
Int J Mol Sci ; 25(16)2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39201718

RÉSUMÉ

Poly (ADP-Ribose) Polymerase (PARP) inhibitors have changed the outcomes and therapeutic strategy for several cancer types. As a targeted therapeutic mainly for patients with BRCA1/2 mutations, PARP inhibitors have commonly been exploited for their capacity to prevent DNA repair. In this review, we discuss the multifaceted roles of PARP-1 and PARP-2 beyond DNA repair, including the impact of PARP-1 on chemokine signalling, immune modulation, and transcriptional regulation of gene expression, particularly in the contexts of angiogenesis and epithelial-to-mesenchymal transition (EMT). We evaluate the pre-clinical role of PARP inhibitors, either as single-agent or combination therapies, to block the metastatic process. Efficacy of PARP inhibitors was demonstrated via DNA repair-dependent and independent mechanisms, including DNA damage, cell migration, invasion, initial colonization at the metastatic site, osteoclastogenesis, and micrometastasis formation. Finally, we summarize the recent clinical advancements of PARP inhibitors in the prevention and progression of distant metastases, with a particular focus on specific metastatic sites and PARP-1 selective inhibitors. Overall, PARP inhibitors have demonstrated great potential in inhibiting the metastatic process, pointing the way for greater use in early cancer settings.


Sujet(s)
Métastase tumorale , Tumeurs , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Poly(ADP-ribose) polymerases , Humains , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Tumeurs/génétique , Poly(ADP-ribose) polymerases/métabolisme , Animaux , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Réparation de l'ADN/effets des médicaments et des substances chimiques
17.
Cancer Discov ; 14(8): 1372-1374, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39091202

RÉSUMÉ

PARP inhibitors (PARPi) are used as a first-line treatment option for cancers with BRCA1/2 mutations, yet a significant number of patients show a limited response to these agents. In the present study, Lei and colleagues demonstrate that PARPi promote increased ferroptosis sensitivity and this can be exploited therapeutically to improve the response to PARPi, marking an important therapeutic concept to exploit ferroptosis-based strategies in clinical settings. See related article by Lei et al., p. 1476 (2).


Sujet(s)
Résistance aux médicaments antinéoplasiques , Ferroptose , Fer , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Ferroptose/effets des médicaments et des substances chimiques , Fer/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/métabolisme
18.
EBioMedicine ; 107: 105288, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39180789

RÉSUMÉ

BACKGROUND: We aimed to assess real-world efficacy of the PARP inhibitor, olaparib, in US Veterans with metastatic prostate cancer (mPC) by leveraging the national data repository and evaluate a novel approach to assess treatment efficacy in tumors considered rare or harboring rare mutations. METHODS: Included Veterans had 1) mPC with somatic or germline alterations/mutations in genes involved in homologous recombination repair (HRR), 2) received olaparib monotherapy as well as a novel hormonal therapy/androgen receptor pathway inhibitors (NHT/ARPI), and/or chemotherapy, and 3) estimable rates of tumor growth (g-rate) using PSA values obtained while receiving treatment. Previous work has shown an excellent inverse correlation of g-rate with survival. Using g-rate, we determined tumor doubling time (DT) and DT ratios (DT on olaparib/DT on prior medication). We postulated that a DT ratio≥ 1 was associated with benefit. FINDINGS: We identified 139 Veterans, including 42 Black males with tumors harboring mutations/alterations in HRR genes who received olaparib: BRCA2 (50), ATM (32), BRCA1 (10), other mutations (47). 62/139 (45%) of all and 21/42 (50%) of Black Veterans had DT ratios ≥1, including 31, 10, 2, and 19 with BRCA2, ATM, BRCA1, and other mutations, respectively (p = 0.006). Median survival with DT ratios ≥1 was superior, being 24.5 vs. 11.4 months for DT ratio <1 (p = 0.01, HR 0.50, 95% CI 0.29-0.85). Benefit from olaparib, defined as DT ratio ≥1, was not observed for germline status, starting PSA value, number of prior therapies, or immediate prior therapy. Compared to matched cohorts, tumors in the olaparib cohort had shorter DTs with enzalutamide in first line (367 vs. 884 days; p = 0.0043). INTERPRETATION: Using equations indifferent to timing of assessments ideal for real-world efficacy analyses, we showed DT ratio ≥1 representing slower tumor growth on olaparib relative to the prior therapy correlates with improved survival. Olaparib efficacy in Veterans with mPC harboring mutations/alterations in HRR genes emulates clinical trial results. Black men had comparable results. Compared to matched cohorts, in first line, enzalutamide was less efficacious in tumors harboring mutations/alterations in HRR genes. FUNDING: American Society of Clinical Oncology Conquer Cancer Foundation (ASCO CCF), the Blavatnik Family Foundation and the Prostate Cancer Foundation (PCF).


Sujet(s)
Phtalazines , Pipérazines , Inhibiteurs de poly(ADP-ribose) polymérases , Tumeurs de la prostate , Anciens combattants , Mâle , Humains , Phtalazines/usage thérapeutique , Pipérazines/usage thérapeutique , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/mortalité , Sujet âgé , Adulte d'âge moyen , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , États-Unis , Résultat thérapeutique , Métastase tumorale , Mutation , Sujet âgé de 80 ans ou plus , Antinéoplasiques/usage thérapeutique
19.
Nat Commun ; 15(1): 6676, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39107288

RÉSUMÉ

53BP1 nucleates the anti-end resection machinery at DNA double-strand breaks, thereby countering BRCA1 activity. Loss of 53BP1 leads to DNA end processing and homologous recombination in BRCA1-deficient cells. Consequently, BRCA1-mutant tumors, typically sensitive to PARP inhibitors (PARPi), become resistant in the absence of 53BP1. Here, we demonstrate that the 'leaky' DNA end resection in the absence of 53BP1 results in increased micronuclei and cytoplasmic double-stranded DNA, leading to activation of the cGAS-STING pathway and pro-inflammatory signaling. This enhances CD8+ T cell infiltration, activates macrophages and natural killer cells, and impedes tumor growth. Loss of 53BP1 correlates with a response to immune checkpoint blockade (ICB) and improved overall survival. Immunohistochemical assessment of 53BP1 in two malignancies, high grade serous ovarian cancer and pancreatic ductal adenocarcinoma, which are refractory to ICBs, reveals that lower 53BP1 levels correlate with an increased adaptive and innate immune response. Finally, BRCA1-deficient tumors that develop resistance to PARPi due to the loss of 53BP1 are susceptible to ICB. Therefore, we conclude that 53BP1 is critical for tumor immunogenicity and underpins the response to ICB. Our results support including 53BP1 expression as an exploratory biomarker in ICB trials for malignancies typically refractory to immunotherapy.


Sujet(s)
Protéines membranaires , Nucleotidyltransferases , Tumeurs de l'ovaire , Tumeurs du pancréas , Protéine-1 liant le suppresseur de tumeur p53 , Protéine-1 liant le suppresseur de tumeur p53/métabolisme , Protéine-1 liant le suppresseur de tumeur p53/génétique , Femelle , Nucleotidyltransferases/métabolisme , Nucleotidyltransferases/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Humains , Animaux , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/métabolisme , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Souris , Lignée cellulaire tumorale , Cassures double-brin de l'ADN , Protéine BRCA1/métabolisme , Protéine BRCA1/génétique , Transduction du signal , Lymphocytes T CD8+/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Souris de lignée C57BL , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Souris knockout , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Immunité innée
20.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39125873

RÉSUMÉ

The current focus of ovarian cancer (OC) research is the improvement of treatment options through maximising drug effectiveness. OC remains the fifth leading cause of cancer-induced mortality in women worldwide. In recent years, nanotechnology has revolutionised drug delivery systems. Nanoparticles may be utilised as carriers in gene therapy or to overcome the problem of drug resistance in tumours by limiting the number of free drugs in circulation and thereby minimising undesired adverse effects. Cell surface receptors, such as human epidermal growth factor 2 (HER2), folic acid (FA) receptors, CD44 (also referred to as homing cell adhesion molecule, HCAM), and vascular endothelial growth factor (VEGF) are highly expressed in ovarian cancer cells. Generation of active targeting nanoparticles involves modification with ligands that recognise cell surface receptors and thereby promote internalisation by cancer cells. Several poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi) are currently used for the treatment of high-grade serous ovarian carcinomas (HGSOC) or platinum-sensitive relapsed OC. However, PARP resistance and poor drug bioavailability are common challenges, highlighting the urgent need to develop novel, effective strategies for ovarian cancer treatment. This review evaluates the utility of nanoparticles in ovarian cancer therapy, with a specific focus on targeted approaches and the use of PARPi nanocarriers to optimise treatment outcomes.


Sujet(s)
Nanoparticules , Tumeurs de l'ovaire , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Femelle , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/métabolisme , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/administration et posologie , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Nanoparticules/composition chimique , Vecteurs de médicaments/composition chimique , Systèmes de délivrance de médicaments/méthodes , Antinéoplasiques/administration et posologie , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Animaux
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE