Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 53
Filtrer
1.
Cancer Res Commun ; 4(9): 2415-2426, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39177285

RÉSUMÉ

PURPOSE: In preclinical models, glucocorticoid receptor (GR) signaling drives resistance to taxane chemotherapy in multiple solid tumors via upregulation of antiapoptotic pathways. ORIC-101 is a potent and selective GR antagonist that was investigated in combination with taxane chemotherapy as an anticancer regimen preclinically and in a phase 1 clinical trial. PATIENTS AND METHODS: The ability of ORIC-101 to reverse taxane resistance was assessed in cell lines and xenograft models, and a phase 1 study (NCT03928314) was conducted in patients with advanced solid tumors to determine the dose, safety, and antitumor activity of ORIC-101 with nab-paclitaxel. RESULTS: ORIC-101 reversed chemoprotection induced by glucocorticoids in vitro and achieved tumor regressions when combined with paclitaxel in both taxane-naïve and -resistant xenograft models. In the phase 1 study, 21 patients were treated in dose escalation and 62 patients were treated in dose expansion. All patients in dose expansion had previously progressed on a taxane-based regimen. In dose escalation, five objective responses were observed. A preplanned futility analysis in dose expansion showed a 3.2% (95% confidence interval, 0.4-11.2) objective response rate with a median progression-free survival of 2 months (95% confidence interval, 1.8-2.8) across all four cohorts, leading to study termination. Pharmacodynamic analysis of tissue and plasma showed GR pathway downregulation in most patients in cycle 1. CONCLUSIONS: ORIC-101 with nab-paclitaxel showed limited clinical activity in taxane-resistant solid tumors. Despite clear inhibition of GR pathway signaling, the insufficient clinical signal underscores the challenges of targeting a single resistance pathway when multiple mechanisms of resistance may be in play. SIGNIFICANCE: Glucocorticoid receptor (GR) upregulation is a mechanism of resistance to taxane chemotherapy in preclinical cancer models. ORIC-101 is a small molecule GR inhibitor. In this phase 1 study, ORIC-101 plus nab-paclitaxel did not show meaningful clinical benefit in patients who previously progressed on taxanes despite successful GR pathway downregulation.


Sujet(s)
Albumines , Protocoles de polychimiothérapie antinéoplasique , Tumeurs , Paclitaxel , Récepteurs aux glucocorticoïdes , Humains , Paclitaxel/usage thérapeutique , Paclitaxel/administration et posologie , Paclitaxel/pharmacologie , Femelle , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Récepteurs aux glucocorticoïdes/antagonistes et inhibiteurs , Récepteurs aux glucocorticoïdes/métabolisme , Mâle , Adulte d'âge moyen , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Sujet âgé , Albumines/administration et posologie , Albumines/usage thérapeutique , Albumines/pharmacologie , Animaux , Adulte , Souris , Tests d'activité antitumorale sur modèle de xénogreffe , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale
3.
Clin Cancer Res ; 30(6): 1111-1120, 2024 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-38226958

RÉSUMÉ

PURPOSE: Increased glucocorticoid receptor (GR) signaling is a proposed compensatory mechanism of resistance to androgen receptor (AR) inhibition in metastatic castration-resistant prostate cancer (mCRPC). ORIC-101 is a potent and selective orally-bioavailable GR antagonist. PATIENTS AND METHODS: Safety, pharmacokinetic/pharmacodynamic, and antitumor activity of ORIC-101 in combination with enzalutamide were studied in patients with mCRPC progressing on enzalutamide. ORIC-101 doses ranging from 80 to 240 mg once daily were tested in combination with enzalutamide 160 mg once daily. Pharmacokinetics/pharmacodynamics was assessed after a single dose and at steady state. Disease control rate (DCR) at 12 weeks was evaluated at the recommended phase 2 dose (RP2D). RESULTS: A total of 41 patients were enrolled. There were no dose-limiting toxicities and the RP2D was selected as 240 mg of ORIC-101 and 160 mg of enzalutamide daily. At the RP2D, the most common treatment-related adverse events were fatigue (38.7%), nausea (29.0%), decreased appetite (19.4%), and constipation (12.9%). Pharmacokinetic/pharmacodynamic data confirmed ORIC-101 achieved exposures necessary for GR target engagement. Overall, for 31 patients treated at the RP2D, there was insufficient clinical benefit based on DCR (25.8%; 80% confidence interval: 15.65-38.52) which did not meet the prespecified target rate, leading to termination of the study. Exploratory subgroup analyses based on baseline GR expression, presence of AR resistance variants, and molecular features of aggressive variant prostate cancer suggested possible benefit in patients with high GR expression and no other resistance markers, although this would require confirmation. CONCLUSIONS: Although the combination of ORIC-101 and enzalutamide demonstrated an acceptable tolerability profile, GR target inhibition with ORIC-101 did not produce clinical benefit in men with metastatic prostate cancer resistant to enzalutamide.


Sujet(s)
Tumeurs prostatiques résistantes à la castration , Mâle , Humains , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Récepteurs aux glucocorticoïdes , 3-Phényl-2-thiohydantoïne , Benzamides/usage thérapeutique , Nitriles/usage thérapeutique , Antinéoplasiques hormonaux/usage thérapeutique
4.
Cancer Res Commun ; 3(9): 1788-1799, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37691854

RÉSUMÉ

The FOXA1 pioneer factor is an essential mediator of steroid receptor function in multiple hormone-dependent cancers, including breast and prostate cancers, enabling nuclear receptors such as estrogen receptor (ER) and androgen receptor (AR) to activate lineage-specific growth programs. FOXA1 is also highly expressed in non-small cell lung cancer (NSCLC), but whether and how it regulates tumor growth in this context is not known. Analyzing data from loss-of-function screens, we identified a subset of NSCLC tumor lines where proliferation is FOXA1 dependent. Using rapid immunoprecipitation and mass spectrometry of endogenous protein, we identified chromatin-localized interactions between FOXA1 and glucocorticoid receptor (GR) in these tumor cells. Knockdown of GR inhibited proliferation of FOXA1-dependent, but not FOXA1-independent NSCLC cells. In these FOXA1-dependent models, FOXA1 and GR cooperate to regulate gene targets involved in EGF signaling and G1-S cell-cycle progression. To investigate the therapeutic potential for targeting this complex, we examined the effects of highly selective inhibitors of the GR ligand-binding pocket and found that GR antagonism with ORIC-101 suppressed FOXA1/GR target expression, activation of EGF signaling, entry into the S-phase, and attendant proliferation in vitro and in vivo. Taken together, our findings point to a subset of NSCLCs harboring a dependence on the FOXA1/GR growth program and provide rationale for its therapeutic targeting. Significance: NSCLC is the leading cause of cancer deaths worldwide. There is a need to identify novel druggable dependencies. We identify a subset of NSCLCs dependent on FOXA1-GR and sensitive to GR antagonism.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Facteur nucléaire hépatocytaire HNF-3 alpha , Tumeurs du poumon , Récepteurs aux glucocorticoïdes , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Facteur de croissance épidermique , Tumeurs du poumon/traitement médicamenteux , Récepteurs aux glucocorticoïdes/génétique , Facteur nucléaire hépatocytaire HNF-3 alpha/génétique
5.
iScience ; 25(12): 105712, 2022 Dec 22.
Article de Anglais | MEDLINE | ID: mdl-36582483

RÉSUMÉ

Here, we have developed an automated image processing algorithm for segmenting lungs and individual lung tumors in in vivo micro-computed tomography (micro-CT) scans of mouse models of non-small cell lung cancer and lung fibrosis. Over 3000 scans acquired across multiple studies were used to train/validate a 3D U-net lung segmentation model and a Support Vector Machine (SVM) classifier to segment individual lung tumors. The U-net lung segmentation algorithm can be used to estimate changes in soft tissue volume within lungs (primarily tumors and blood vessels), whereas the trained SVM is able to discriminate between tumors and blood vessels and identify individual tumors. The trained segmentation algorithms (1) significantly reduce time required for lung and tumor segmentation, (2) reduce bias and error associated with manual image segmentation, and (3) facilitate identification of individual lung tumors and objective assessment of changes in lung and individual tumor volumes under different experimental conditions.

6.
Elife ; 112022 08 19.
Article de Anglais | MEDLINE | ID: mdl-35983994

RÉSUMÉ

Lung development, integrity and repair rely on precise Wnt signaling, which is corrupted in diverse diseases, including cancer. Here, we discover that EHMT2 methyltransferase regulates Wnt signaling in the lung by controlling the transcriptional activity of chromatin-bound ß-catenin, through a non-histone substrate in mouse lung. Inhibition of EHMT2 induces transcriptional, morphologic, and molecular changes consistent with alveolar type 2 (AT2) lineage commitment. Mechanistically, EHMT2 activity functions to support regenerative properties of KrasG12D tumors and normal AT2 cells-the predominant cell of origin of this cancer. Consequently, EHMT2 inhibition prevents KrasG12D lung adenocarcinoma (LUAD) tumor formation and propagation and disrupts normal AT2 cell differentiation. Consistent with these findings, low gene EHMT2 expression in human LUAD correlates with enhanced AT2 gene expression and improved prognosis. These data reveal EHMT2 as a critical regulator of Wnt signaling, implicating Ehmt2 as a potential target in lung cancer and other AT2-mediated lung pathologies.


Sujet(s)
Adénocarcinome pulmonaire , Tumeurs du poumon , Adénocarcinome pulmonaire/génétique , Animaux , Gènes ras , Antigènes d'histocompatibilité/génétique , Histone-lysine N-methyltransferase/génétique , Histone-lysine N-methyltransferase/métabolisme , Humains , Poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Methyltransferases/métabolisme , Souris , Protéines proto-oncogènes p21(ras)/métabolisme
7.
Cell Rep ; 38(6): 110351, 2022 02 08.
Article de Anglais | MEDLINE | ID: mdl-35139374

RÉSUMÉ

KRAS, which is mutated in ∼30% of all cancers, activates the RAF-MEK-ERK signaling cascade. CRAF is required for growth of KRAS mutant lung tumors, but the requirement for CRAF kinase activity is unknown. Here, we show that subsets of KRAS mutant tumors are dependent on CRAF for growth. Kinase-dead but not dimer-defective CRAF rescues growth inhibition, suggesting that dimerization but not kinase activity is required. Quantitative proteomics demonstrates increased levels of CRAF:ARAF dimers in KRAS mutant cells, and depletion of both CRAF and ARAF rescues the CRAF-loss phenotype. Mechanistically, CRAF depletion causes sustained ERK activation and induction of cell-cycle arrest, while treatment with low-dose MEK or ERK inhibitor rescues the CRAF-loss phenotype. Our studies highlight the role of CRAF in regulating MAPK signal intensity to promote tumorigenesis downstream of mutant KRAS and suggest that disrupting CRAF dimerization or degrading CRAF may have therapeutic benefit.


Sujet(s)
Carcinogenèse/métabolisme , Dimérisation , Protéines proto-oncogènes p21(ras)/métabolisme , Animaux , Carcinogenèse/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Humains , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Souris , Phosphorylation/physiologie , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes B-raf/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/physiologie , Protéines G ras/génétique
8.
Nat Rev Drug Discov ; 21(2): 99-114, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34702990

RÉSUMÉ

Recent progress in understanding the molecular basis of cellular processes, identification of promising therapeutic targets and evolution of the regulatory landscape makes this an exciting and unprecedented time to be in the field of oncology drug development. However, high costs, long development timelines and steep rates of attrition continue to afflict the drug development process. Lack of predictive preclinical models is considered one of the key reasons for the high rate of attrition in oncology. Generating meaningful and predictive results preclinically requires a firm grasp of the relevant biological questions and alignment of the model systems that mirror the patient context. In doing so, the ability to conduct both forward translation, the process of implementing basic research discoveries into practice, as well as reverse translation, the process of elucidating the mechanistic basis of clinical observations, greatly enhances our ability to develop effective anticancer treatments. In this Review, we outline issues in preclinical-to-clinical translatability of molecularly targeted cancer therapies, present concepts and examples of successful reverse translation, and highlight the need to better align tumour biology in patients with preclinical model systems including tracking of strengths and weaknesses of preclinical models throughout programme development.


Sujet(s)
Antinéoplasiques/pharmacologie , Marqueurs biologiques tumoraux/métabolisme , Développement de médicament , Évaluation préclinique de médicament/méthodes , Thérapie moléculaire ciblée , Tumeurs/traitement médicamenteux , Animaux , Marqueurs biologiques tumoraux/génétique , Humains , Tumeurs/génétique , Tumeurs/métabolisme , Tumeurs/anatomopathologie
9.
Nat Commun ; 12(1): 6967, 2021 11 29.
Article de Anglais | MEDLINE | ID: mdl-34845227

RÉSUMÉ

Breast cancer is now globally the most frequent cancer and leading cause of women's death. Two thirds of breast cancers express the luminal estrogen receptor-positive (ERα + ) phenotype that is initially responsive to antihormonal therapies, but drug resistance emerges. A major barrier to the understanding of the ERα-pathway biology and therapeutic discoveries is the restricted repertoire of luminal ERα + breast cancer models. The ERα + phenotype is not stable in cultured cells for reasons not fully understood. We examine 400 patient-derived breast epithelial and breast cancer explant cultures (PDECs) grown in various three-dimensional matrix scaffolds, finding that ERα is primarily regulated by the matrix stiffness. Matrix stiffness upregulates the ERα signaling via stress-mediated p38 activation and H3K27me3-mediated epigenetic regulation. The finding that the matrix stiffness is a central cue to the ERα phenotype reveals a mechanobiological component in breast tissue hormonal signaling and enables the development of novel therapeutic interventions. Subject terms: ER-positive (ER + ), breast cancer, ex vivo model, preclinical model, PDEC, stiffness, p38 SAPK.


Sujet(s)
Tumeurs du sein/génétique , Récepteur alpha des oestrogènes/génétique , Mécanotransduction cellulaire/génétique , Transcriptome , p38 Mitogen-Activated Protein Kinases/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Études cas-témoins , Lignée cellulaire tumorale , Cinnamates/pharmacologie , Collagène/composition chimique , Collagène/pharmacologie , Association médicamenteuse , Protéine-2 homologue de l'activateur de Zeste/génétique , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Oestradiol/pharmacologie , Récepteur alpha des oestrogènes/métabolisme , Femelle , Fulvestrant/pharmacologie , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Histone/génétique , Histone/métabolisme , Humains , Indazoles/pharmacologie , Laminine/composition chimique , Laminine/pharmacologie , Glandes mammaires humaines/effets des médicaments et des substances chimiques , Glandes mammaires humaines/métabolisme , Glandes mammaires humaines/anatomopathologie , Phénotype , Protéoglycanes/composition chimique , Protéoglycanes/pharmacologie , Tamoxifène/pharmacologie , Techniques de culture de tissus , p38 Mitogen-Activated Protein Kinases/métabolisme
10.
Clin Cancer Res ; 27(4): 1162-1173, 2021 02 15.
Article de Anglais | MEDLINE | ID: mdl-33023953

RÉSUMÉ

PURPOSE: Lung adenocarcinomas comprise the largest fraction of non-small cell lung cancer, which is the leading cause of cancer-related deaths. Seventy-five percent of adenocarcinomas lack targeted therapies because of scarcity of druggable drivers. Here, we classified tumors on the basis of signaling similarities and discovered subgroups within this unmet patient population. EXPERIMENTAL DESIGN: We leveraged transcriptional data from >800 early- and advanced-stage patients. RESULTS: We identified three robust subtypes dubbed mucinous, proliferative, and mesenchymal with respective pathway phenotypes. These transcriptional states lack discrete and causative mutational etiology as evidenced by similarly distributed oncogenic drivers, including KRAS and EGFR. The subtypes capture heterogeneity even among tumors lacking known oncogenic drivers. Paired multi-regional intratumoral biopsies demonstrated unified subtypes despite divergently evolved prooncogenic mutations, indicating subtype stability during selective pressure. Heterogeneity among in vitro and in vivo preclinical models is expounded by the human lung adenocarcinoma subtypes and can be leveraged to discover subtype-specific vulnerabilities. As proof of concept, we identified differential subtype response to MEK pathway inhibition in a chemical library screen of 89 lung cancer cell lines, which reproduces across model systems and a clinical trial. CONCLUSIONS: Our findings support forward translational relevance of transcriptional subtypes, where further exploration therein may improve lung adenocarcinoma treatment.See related commentary by Skoulidis, p. 913.


Sujet(s)
Adénocarcinome pulmonaire/traitement médicamenteux , Marqueurs biologiques tumoraux/génétique , Tumeurs du poumon/traitement médicamenteux , Mitogen-Activated Protein Kinase Kinases/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/usage thérapeutique , Adénocarcinome pulmonaire/diagnostic , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Animaux , Lignée cellulaire tumorale , Essais cliniques comme sujet , Jeux de données comme sujet , Femelle , Hétérogénéité génétique , Humains , Tumeurs du poumon/diagnostic , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Mâle , Souris , Stadification tumorale , Inhibiteurs de protéines kinases/pharmacologie , RNA-Seq , Transcriptome/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
11.
J Med Chem ; 63(18): 10433-10459, 2020 09 24.
Article de Anglais | MEDLINE | ID: mdl-32865411

RÉSUMÉ

The adenosinergic pathway represents an attractive new therapeutic approach in cancer immunotherapy. In this pathway, ecto-5-nucleotidase CD73 has the unique function of regulating production of immunosuppressive adenosine (ADO) through the hydrolysis of AMP. CD73 is overexpressed in many cancers, resulting in elevated levels of ADO that correspond to poor patient prognosis. Therefore, reducing the level of ADO via inhibition of CD73 is a potential strategy for treating cancers. Based on the binding mode of adenosine 5'-(α,ß-methylene)diphosphate (AOPCP) with human CD73, we designed a series of novel monophosphonate small-molecule CD73 inhibitors. Among them, OP-5244 (35) proved to be a highly potent and orally bioavailable CD73 inhibitor. In preclinical studies, 35 completely inhibited ADO production in both human cancer cells and CD8+ T cells. Furthermore, 35 lowered the ratio of ADO/AMP significantly and reversed immunosuppression in mouse models, indicating its potential as an in vivo tool compound for further development.


Sujet(s)
5'-Nucleotidase/antagonistes et inhibiteurs , Adénosine/antagonistes et inhibiteurs , Antienzymes/pharmacologie , Facteurs immunologiques/pharmacologie , Nucléosides/pharmacologie , Phosphonates/pharmacologie , Administration par voie orale , Animaux , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Chiens , Antienzymes/administration et posologie , Antienzymes/synthèse chimique , Antienzymes/pharmacocinétique , Protéines liées au GPI/antagonistes et inhibiteurs , Humains , Facteurs immunologiques/administration et posologie , Facteurs immunologiques/synthèse chimique , Facteurs immunologiques/pharmacocinétique , Macaca fascicularis , Souris de lignée BALB C , Structure moléculaire , Nucléosides/administration et posologie , Nucléosides/synthèse chimique , Nucléosides/pharmacocinétique , Phosphonates/administration et posologie , Phosphonates/synthèse chimique , Phosphonates/pharmacocinétique , Rats , Stéréoisomérie , Relation structure-activité
12.
JCI Insight ; 5(7)2020 04 09.
Article de Anglais | MEDLINE | ID: mdl-32271166

RÉSUMÉ

Systemic cytokine release and on-target/off-tumor toxicity to normal tissues are the main adverse effects limiting the clinical utility of T cell-redirecting therapies. This study was designed to determine how binding affinity for CD3 and tumor target HER2 impact the efficacy and nonclinical safety of anti-HER2/CD3 T cell-dependent antibodies (TDBs). Affinity was found to be a major determinant for the overall tolerability. Higher affinity for CD3 associated with rapidly elevated peripheral cytokine concentrations, weight loss in mice, and poor tolerability in cynomolgus monkeys. A TDB with lower CD3 affinity was better tolerated in cynomolgus monkeys compared with a higher CD3-affinity TDB. In contrast to tolerability, T cell binding affinity had only limited impact on in vitro and in vivo antitumor activity. High affinity for HER2 was critical for the tumor-killing activity of anti-HER2/CD3 TDBs, but higher HER2 affinity also associated with a more severe toxicity profile, including cytokine release and damage to HER2-expressing tissues. The tolerability of the anti-HER2/CD3 was improved by implementing a dose-fractionation strategy. Fine-tuning the affinities for both the tumor target and CD3 is likely a valuable strategy for achieving maximal therapeutic index of CD3 bispecific antibodies.


Sujet(s)
Anticorps bispécifiques/immunologie , Affinité des anticorps , Antinéoplasiques immunologiques/immunologie , Récepteur ErbB-2/immunologie , Animaux , Anticorps bispécifiques/composition chimique , Antinéoplasiques immunologiques/composition chimique , Antigènes CD3/composition chimique , Cellules CHO , Cricetulus , Évaluation préclinique de médicament , Humains , Macaca fascicularis , Récepteur ErbB-2/composition chimique
14.
Cancer Discov ; 10(2): 232-253, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31699795

RÉSUMÉ

With only a fraction of patients responding to cancer immunotherapy, a better understanding of the entire tumor microenvironment is needed. Using single-cell transcriptomics, we chart the fibroblastic landscape during pancreatic ductal adenocarcinoma (PDAC) progression in animal models. We identify a population of carcinoma-associated fibroblasts (CAF) that are programmed by TGFß and express the leucine-rich repeat containing 15 (LRRC15) protein. These LRRC15+ CAFs surround tumor islets and are absent from normal pancreatic tissue. The presence of LRRC15+ CAFs in human patients was confirmed in >80,000 single cells from 22 patients with PDAC as well as by using IHC on samples from 70 patients. Furthermore, immunotherapy clinical trials comprising more than 600 patients across six cancer types revealed elevated levels of the LRRC15+ CAF signature correlated with poor response to anti-PD-L1 therapy. This work has important implications for targeting nonimmune elements of the tumor microenvironment to boost responses of patients with cancer to immune checkpoint blockade therapy. SIGNIFICANCE: This study describes the single-cell landscape of CAFs in pancreatic cancer during in vivo tumor evolution. A TGFß-driven, LRRC15+ CAF lineage is associated with poor outcome in immunotherapy trial data comprising multiple solid-tumor entities and represents a target for combinatorial therapy.This article is highlighted in the In This Issue feature, p. 161.


Sujet(s)
Fibroblastes associés au cancer/immunologie , Carcinome du canal pancréatique/traitement médicamenteux , Résistance aux médicaments antinéoplasiques/génétique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Protéines membranaires/métabolisme , Myofibroblastes/immunologie , Tumeurs du pancréas/traitement médicamenteux , Animaux , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques , Fibroblastes associés au cancer/métabolisme , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/immunologie , Lignée cellulaire tumorale , Lignage cellulaire/génétique , Lignage cellulaire/immunologie , Essais cliniques comme sujet , Biologie informatique , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/immunologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/immunologie , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Souris , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/métabolisme , Tumeurs du pancréas/génétique , Tumeurs du pancréas/immunologie , RNA-Seq , Analyse sur cellule unique , Facteur de croissance transformant bêta/métabolisme , Résultat thérapeutique , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie
15.
Cell Death Differ ; 27(1): 161-175, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31101885

RÉSUMÉ

The kinase RIP1 acts in multiple signaling pathways to regulate inflammatory responses and it can trigger both apoptosis and necroptosis. Its kinase activity has been implicated in a range of inflammatory, neurodegenerative, and oncogenic diseases. Here, we explore the effect of inhibiting RIP1 genetically, using knock-in mice that express catalytically inactive RIP1 D138N, or pharmacologically, using the murine-potent inhibitor GNE684. Inhibition of RIP1 reduced collagen antibody-induced arthritis, and prevented skin inflammation caused by mutation of Sharpin, or colitis caused by deletion of Nemo from intestinal epithelial cells. Conversely, inhibition of RIP1 had no effect on tumor growth or survival in pancreatic tumor models driven by mutant Kras, nor did it reduce lung metastases in a B16 melanoma model. Collectively, our data emphasize a role for the kinase activity of RIP1 in certain inflammatory disease models, but question its relevance to tumor progression and metastases.


Sujet(s)
Inflammation/enzymologie , Tumeurs/enzymologie , Receptor-Interacting Protein Serine-Threonine Kinases/antagonistes et inhibiteurs , Animaux , Arthrite/enzymologie , Mort cellulaire , Lignée cellulaire , Lignée cellulaire tumorale , Colite/étiologie , Colite/prévention et contrôle , Dermatite/enzymologie , Femelle , Techniques de knock-in de gènes , Humains , Iléite/étiologie , Iléite/prévention et contrôle , Protéines et peptides de signalisation intracellulaire/génétique , Mâle , Mélanome expérimental/anatomopathologie , Souris , Métastase tumorale , Tumeurs du pancréas/anatomopathologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Rats , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Receptor-Interacting Protein Serine-Threonine Kinases/physiologie
16.
Sci Transl Med ; 11(508)2019 09 04.
Article de Anglais | MEDLINE | ID: mdl-31484792

RÉSUMÉ

T cell-retargeting therapies have transformed the therapeutic landscape of oncology. Regardless of the modality, T cell activating therapies are commonly accompanied by systemic cytokine release, which can progress to deadly cytokine release syndrome (CRS). Because of incomplete mechanistic understanding of the relationship between T cell activation and systemic cytokine release, optimal toxicity management that retains full therapeutic potential remains unclear. Here, we report the cell type-specific cellular mechanisms that link CD3 bispecific antibody-mediated killing to toxic cytokine release. The immunologic cascade is initiated by T cell triggering, whereas monocytes and macrophages are the primary source of systemic toxic cytokine release. We demonstrate that T cell-generated tumor necrosis factor-α (TNF-α) is the primary mechanism mediating monocyte activation and systemic cytokine release after CD3 bispecific treatment. Prevention of TNF-α release is sufficient to impair systemic release of monocyte cytokines without affecting antitumor efficacy. Systemic cytokine release is only observed upon initial exposure to CD3 bispecific antibody not subsequent doses, indicating a biological distinction between doses. Despite impaired cytokine release after second exposure, T cell cytotoxicity remained unaffected, demonstrating that cytolytic activity of T cells can be achieved in the absence of cytokine release. The mechanistic uncoupling of toxic cytokines and T cell cytolytic activity in the context of CD3 bispecifics provides a biological rationale to clinically explore preventative treatment approaches to mitigate toxicity.


Sujet(s)
Anticorps bispécifiques/immunologie , Antigènes CD3/immunologie , Cytokines/métabolisme , Cytotoxicité immunologique , Lymphocytes T cytotoxiques/immunologie , Animaux , Humains , Macrophages/métabolisme , Souris transgéniques , Monocytes/métabolisme , Récepteur ErbB-2/métabolisme , Facteur de nécrose tumorale alpha/métabolisme
17.
Immunity ; 51(1): 119-130.e5, 2019 07 16.
Article de Anglais | MEDLINE | ID: mdl-31231034

RÉSUMÉ

Tissue-resident macrophages require specific milieus for the maintenance of defining gene-expression programs. Expression of the transcription factor GATA6 is required for the homeostasis, function and localization of peritoneal cavity-resident macrophages. Gata6 expression is maintained in a non-cell autonomous manner and is elicited by the vitamin A metabolite, retinoic acid. Here, we found that the GATA6 transcriptional program is a common feature of macrophages residing in all visceral body cavities. Retinoic acid-dependent and -independent hallmark genes of GATA6+ macrophages were induced by mesothelial and fibroblastic stromal cells that express the transcription factor Wilms' Tumor 1 (WT1), which drives the expression of two rate-limiting enzymes in retinol metabolism. Depletion of Wt1+ stromal cells reduced the frequency of GATA6+ macrophages in the peritoneal, pleural and pericardial cavities. Thus, Wt1+ mesothelial and fibroblastic stromal cells constitute essential niche components supporting the tissue-specifying transcriptional landscape and homeostasis of cavity-resident macrophages.


Sujet(s)
Facteur de transcription GATA-6/métabolisme , Macrophages/physiologie , Péricarde/immunologie , Cavité péritonéale/physiologie , Cavité pleurale/immunologie , Protéines de répression/métabolisme , Cellules stromales/physiologie , Animaux , Différenciation cellulaire , Cellules cultivées , Facteur de transcription GATA-6/génétique , Homéostasie , Souris , Souris de lignée C57BL , Souris transgéniques , Protéines de répression/génétique , Trétinoïne/métabolisme , Protéines WT1
18.
Cell Death Differ ; 26(11): 2416-2429, 2019 Nov.
Article de Anglais | MEDLINE | ID: mdl-30824837

RÉSUMÉ

Emerging research suggests that multiple tumor compartments can influence treatment responsiveness and relapse, yet the search for therapeutic resistance mechanisms remains largely focused on acquired genomic alterations in cancer cells. Here we show how treatment-induced changes occur in multiple tumor compartments during tumor relapse and can reduce benefit of follow-on therapies. By using serial biopsies, next-generation sequencing, and single-cell transcriptomics, we tracked the evolution of multiple cellular compartments within individual lesions during first-line treatment response, relapse, and second-line therapeutic interventions in an autochthonous model of melanoma. We discovered that although treatment-relapsed tumors remained genetically stable, they converged on a shared resistance phenotype characterized by dramatic changes in tumor cell differentiation state, immune infiltration, and extracellular matrix (ECM) composition. Similar alterations in tumor cell differentiation were also observed in more than half of our treatment-relapsed patient tumors. Tumor cell-state changes were coincident with ECM remodeling and increased tumor stiffness, which alone was sufficient to alter tumor cell fate and reduce treatment responses in melanoma cell lines in vitro. Despite the absence of acquired mutations in the targeted pathway, resistant tumors showed significantly decreased responsiveness to second-line therapy intervention within the same pathway. The ability to preclinically model relapse and refractory settings-while capturing dynamics within and crosstalk between all relevant tumor compartments-provides a unique opportunity to better design and sequence appropriate clinical interventions.


Sujet(s)
Antinéoplasiques/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Matrice extracellulaire/anatomopathologie , Mélanome/traitement médicamenteux , Mélanome/anatomopathologie , Animaux , Azétidines/pharmacologie , Marqueurs biologiques tumoraux/génétique , Lignée cellulaire tumorale , Variations de nombre de copies de segment d'ADN/génétique , Résistance aux médicaments antinéoplasiques/physiologie , Femelle , Séquençage nucléotidique à haut débit , Humains , Mâle , Mélanome/génétique , Souris , Souris de lignée C57BL , Souris transgéniques , Mutation/génétique , Pipéridines/pharmacologie , Protéines proto-oncogènes B-raf/génétique , Vémurafénib/pharmacologie ,
19.
Nat Commun ; 9(1): 2992, 2018 07 31.
Article de Anglais | MEDLINE | ID: mdl-30065304

RÉSUMÉ

Delta ligands regulate Notch signaling in normal intestinal stem cells, while Jagged1 activates Notch in intestinal adenomas carrying active ß-catenin. We used the ApcMin/+ mouse model, tumor spheroid cultures, and patient-derived orthoxenografts to address this divergent ligand-dependent Notch function and its implication in disease. We found that intestinal-specific Jag1 deletion or antibody targeting Jag1 prevents tumor initiation in mice. Addiction to Jag1 is concomitant with the absence of Manic Fringe (MFNG) in adenoma cells, and its ectopic expression reverts Jag1 dependence. In 239 human colorectal cancer patient samples, MFNG imposes a negative correlation between Jag1 and Notch, being high Jag1 in the absence of MFNG predictive of poor prognosis. Jag1 antibody treatment reduces patient-derived tumor orthoxenograft growth without affecting normal intestinal mucosa. Our data provide an explanation to Jag1 dependence in cancer, and reveal that Jag1-Notch1 interference provides therapeutic benefit in a subset of colorectal cancer and FAP syndrome patients.


Sujet(s)
Hexosyltransferases/métabolisme , Tumeurs de l'intestin/métabolisme , Tumeurs de l'intestin/anatomopathologie , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéine jagged-1/métabolisme , Protéines membranaires/métabolisme , Protéines/métabolisme , Animaux , Marqueurs biologiques tumoraux/métabolisme , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Prolifération cellulaire , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Glucosyltransferases , Humains , Ligands , Souris , Modèles biologiques , Pronostic , Récepteur Notch1/métabolisme , Transduction du signal , Sphéroïdes de cellules/métabolisme , Sphéroïdes de cellules/anatomopathologie , Cellules souches/métabolisme , Transcription génétique
20.
NPJ Precis Oncol ; 2(1): 7, 2018.
Article de Anglais | MEDLINE | ID: mdl-29872725

RÉSUMÉ

KRAS- and BRAF-mutant tumors are often dependent on MAPK signaling for proliferation and survival and thus sensitive to MAPK pathway inhibitors. However, clinical studies have shown that MEK inhibitors are not uniformly effective in these cancers indicating that mutational status of these oncogenes does not accurately capture MAPK pathway activity. A number of transcripts are regulated by this pathway and are recurrently identified in genome-based MAPK transcriptional signatures. To test whether the transcriptional output of only 10 of these targets could quantify MAPK pathway activity with potential predictive or prognostic clinical utility, we created a MAPK Pathway Activity Score (MPAS) derived from aggregated gene expression. In vitro, MPAS predicted sensitivity to MAPK inhibitors in multiple cell lines, comparable to or better than larger genome-based statistical models. Bridging in vitro studies and clinical samples, median MPAS from a given tumor type correlated with cobimetinib (MEK inhibitor) sensitivity of cancer cell lines originating from the same tissue type. Retrospective analyses of clinical datasets showed that MPAS was associated with the sensitivity of melanomas to vemurafenib (HR: 0.596) and negatively prognostic of overall or progression-free survival in both adjuvant and metastatic CRC (HR: 1.5 and 1.4), adrenal cancer (HR: 1.7), and HER2+ breast cancer (HR: 1.6). MPAS thus demonstrates potential clinical utility that warrants further exploration.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE