Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 30
Filtrer
1.
Cancer Discov ; 13(8): 1789-1801, 2023 08 04.
Article de Anglais | MEDLINE | ID: mdl-37269335

RÉSUMÉ

Rationally targeted therapies have transformed cancer treatment, but many patients develop resistance through bypass signaling pathway activation. PF-07284892 (ARRY-558) is an allosteric SHP2 inhibitor designed to overcome bypass-signaling-mediated resistance when combined with inhibitors of various oncogenic drivers. Activity in this setting was confirmed in diverse tumor models. Patients with ALK fusion-positive lung cancer, BRAFV600E-mutant colorectal cancer, KRASG12D-mutant ovarian cancer, and ROS1 fusion-positive pancreatic cancer who previously developed targeted therapy resistance were treated with PF-07284892 on the first dose level of a first-in-human clinical trial. After progression on PF-07284892 monotherapy, a novel study design allowed the addition of oncogene-directed targeted therapy that had previously failed. Combination therapy led to rapid tumor and circulating tumor DNA (ctDNA) responses and extended the duration of overall clinical benefit. SIGNIFICANCE: PF-07284892-targeted therapy combinations overcame bypass-signaling-mediated resistance in a clinical setting in which neither component was active on its own. This provides proof of concept of the utility of SHP2 inhibitors in overcoming resistance to diverse targeted therapies and provides a paradigm for accelerated testing of novel drug combinations early in clinical development. See related commentary by Hernando-Calvo and Garralda, p. 1762. This article is highlighted in the In This Issue feature, p. 1749.


Sujet(s)
Tumeurs du poumon , Protein-tyrosine kinases , Humains , Protein-tyrosine kinases/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Oncogènes , Soins centrés sur le patient
2.
Nat Commun ; 13(1): 5959, 2022 10 10.
Article de Anglais | MEDLINE | ID: mdl-36216827

RÉSUMÉ

Multigene assays can provide insight into key biological processes and prognostic information to guide development and selection of adjuvant cancer therapy. We report a comprehensive genomic and transcriptomic analysis of tumor samples from 171 patients at high risk for recurrent renal cell carcinoma post nephrectomy from the S-TRAC trial (NCT00375674). We identify gene expression signatures, including STRAC11 (derived from the sunitinib-treated population). The overlap in key elements captured in these gene expression signatures, which include genes representative of the tumor stroma microenvironment, regulatory T cell, and myeloid cells, suggests they are likely to be both prognostic and predictive of the anti-angiogenic effect in the adjuvant setting. These signatures also point to the identification of potential therapeutic targets for development in adjuvant renal cell carcinoma, such as MERTK and TDO2. Finally, our findings suggest that while anti-angiogenic adjuvant therapy might be important, it may not be sufficient to prevent recurrence and that other factors such as immune response and tumor environment may be of greater importance.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Adjuvants immunologiques/usage thérapeutique , Néphrocarcinome/traitement médicamenteux , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Traitement médicamenteux adjuvant , Essais cliniques de phase III comme sujet , Humains , Tumeurs du rein/traitement médicamenteux , Tumeurs du rein/génétique , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/génétique , Sunitinib/usage thérapeutique , Microenvironnement tumoral/génétique , c-Mer Tyrosine kinase
3.
Ann Hematol ; 101(8): 1689-1701, 2022 Aug.
Article de Anglais | MEDLINE | ID: mdl-35488900

RÉSUMÉ

This phase 1b study evaluated glasdegib (100 mg once daily) + azacitidine in adults with newly diagnosed acute myeloid leukemia (AML), higher-risk myelodysplastic syndromes (MDS), or chronic myelomonocytic leukemia (CMML) who were ineligible for intensive chemotherapy. Of 72 patients enrolled, 12 were in a lead-in safety cohort (LIC) and 60 were in the AML and MDS (including CMML) expansion cohorts. In the LIC, the safety profile of glasdegib + azacitidine was determined to be consistent with those of glasdegib or azacitidine alone, with no evidence of drug-drug interaction. In the expansion cohort, the most frequently (≥ 10%) reported non-hematologic Grade ≥ 3 treatment-emergent adverse events were decreased appetite, electrocardiogram QT prolongation, and hypertension in the AML cohort and sepsis, diarrhea, hypotension, pneumonia, and hyperglycemia in the MDS cohort. Overall response rates in the AML and MDS cohorts were 30.0% and 33.3%, respectively; 47.4% and 46.7% of patients who were transfusion dependent at baseline achieved independence. Median overall survival (95% confidence interval) was 9.2 (6.2-14.0) months and 15.8 (9.3-21.9) months, respectively, and response was associated with molecular mutation clearance. Glasdegib + azacitidine in patients with newly diagnosed AML or MDS demonstrated an acceptable safety profile and preliminary evidence of clinical benefits.Trial registration: ClinicalTrials.gov NCT02367456.


Sujet(s)
Leucémie aigüe myéloïde , Syndromes myélodysplasiques , Adulte , Azacitidine/effets indésirables , Benzimidazoles/effets indésirables , Association de médicaments/effets indésirables , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Syndromes myélodysplasiques/traitement médicamenteux , Phénylurées/effets indésirables , Appréciation des risques , Résultat thérapeutique
4.
Nat Med ; 27(12): 2200-2211, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34893775

RÉSUMÉ

In a recent phase 3 randomized trial of 700 patients with advanced urothelial cancer (JAVELIN Bladder 100; NCT02603432 ), avelumab/best supportive care (BSC) significantly prolonged overall survival relative to BSC alone as maintenance therapy after first-line chemotherapy. Exploratory biomarker analyses were performed to identify biological pathways that might affect survival benefit. Tumor molecular profiling by immunohistochemistry, whole-exome sequencing and whole-transcriptome sequencing revealed that avelumab survival benefit was positively associated with PD-L1 expression by tumor cells, tumor mutational burden, APOBEC mutation signatures, expression of genes underlying innate and adaptive immune activity and the number of alleles encoding high-affinity variants of activating Fcγ receptors. Pathways connected to tissue growth and angiogenesis might have been associated with reduced survival benefit. Individual biomarkers did not comprehensively identify patients who could benefit from therapy; however, multi-parameter models incorporating genomic alteration, immune responses and tumor growth showed promising predictive utility. These results characterize the complex biologic pathways underlying survival benefit from immune checkpoint inhibition in advanced urothelial cancer and suggest that multiple biomarkers might be needed to identify patients who would benefit from treatment.


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Antinéoplasiques immunologiques/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Tumeurs de la vessie urinaire/traitement médicamenteux , Humains , Mutation , Tumeurs de la vessie urinaire/anatomopathologie
5.
Cancer Immunol Res ; 9(10): 1141-1157, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34376502

RÉSUMÉ

The use of cytokines for immunotherapy shows clinical efficacy but is frequently accompanied by severe adverse events caused by excessive and systemic immune activation. Here, we set out to address these challenges by engineering a fusion protein of a single, potency-reduced, IL15 mutein and a PD1-specific antibody (anti-PD1-IL15m). This immunocytokine was designed to deliver PD1-mediated, avidity-driven IL2/15 receptor stimulation to PD1+ tumor-infiltrating lymphocytes (TIL) while minimally affecting circulating peripheral natural killer (NK) cells and T cells. Treatment of tumor-bearing mice with a mouse cross-reactive fusion, anti-mPD1-IL15m, demonstrated potent antitumor efficacy without exacerbating body weight loss in B16 and MC38 syngeneic tumor models. Moreover, anti-mPD1-IL15m was more efficacious than an IL15 superagonist, an anti-mPD-1, or the combination thereof in the B16 melanoma model. Mechanistically, anti-PD1-IL15m preferentially targeted CD8+ TILs and single-cell RNA-sequencing analyses revealed that anti-mPD1-IL15m treatment induced the expansion of an exhausted CD8+ TIL cluster with high proliferative capacity and effector-like signatures. Antitumor efficacy of anti-mPD1-IL15m was dependent on CD8+ T cells, as depletion of CD8+ cells resulted in the loss of antitumor activity, whereas depletion of NK cells had little impact on efficacy. The impact of anti-hPD1-IL15m on primary human TILs from patients with cancer was also evaluated. Anti-hPD1-IL15m robustly enhanced the proliferation, activation, and cytotoxicity of CD8+ and CD4+ TILs from human primary cancers in vitro, whereas tumor-derived regulatory T cells were largely unaffected. Taken together, our findings showed that anti-PD1-IL15m exhibits a high translational promise with improved efficacy and safety of IL15 for cancer immunotherapy via targeting PD1+ TILs.See related Spotlight by Felices and Miller, p. 1110.


Sujet(s)
Lymphocytes T CD8+/immunologie , Tumeurs du côlon/thérapie , Immunothérapie , Interleukine-15/usage thérapeutique , Mélanome expérimental/thérapie , Animaux , Lignée cellulaire tumorale , Tumeurs du côlon/immunologie , Modèles animaux de maladie humaine , Humains , Interleukine-15/immunologie , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Mélanome expérimental/immunologie , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Récepteur-1 de mort cellulaire programmée/immunologie , Ingénierie des protéines , Protéines de fusion recombinantes/immunologie , Protéines de fusion recombinantes/usage thérapeutique
6.
Nat Commun ; 11(1): 6175, 2020 12 02.
Article de Anglais | MEDLINE | ID: mdl-33268821

RÉSUMÉ

To elucidate the effects of neoadjuvant chemotherapy (NAC), we conduct whole transcriptome profiling coupled with histopathology analyses of a longitudinal breast cancer cohort of 146 patients including 110 pairs of serial tumor biopsies collected before treatment, after the first cycle of treatment and at the time of surgery. Here, we show that cytotoxic chemotherapies induce dynamic changes in the tumor immune microenvironment that vary by subtype and pathologic response. Just one cycle of treatment induces an immune stimulatory microenvironment harboring more tumor infiltrating lymphocytes (TILs) and up-regulation of inflammatory signatures predictive of response to anti-PD1 therapies while residual tumors are immune suppressed at end-of-treatment compared to the baseline. Increases in TILs and CD8+ T cell proportions in response to NAC are independently associated with pathologic complete response. Further, on-treatment immune response is more predictive of treatment outcome than immune features in paired baseline samples although these are strongly correlated.


Sujet(s)
Antigène CD274/génétique , Tumeurs du sein/traitement médicamenteux , Carcinome canalaire du sein/traitement médicamenteux , Régulation de l'expression des gènes tumoraux , Lymphocytes TIL/effets des médicaments et des substances chimiques , Traitement néoadjuvant/méthodes , Anthracyclines/usage thérapeutique , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Tumeurs du sein/mortalité , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/anatomopathologie , Carcinome canalaire du sein/génétique , Carcinome canalaire du sein/immunologie , Carcinome canalaire du sein/mortalité , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/immunologie , Cyclophosphamide/usage thérapeutique , Survie sans rechute , Docetaxel/usage thérapeutique , Récepteur alpha des oestrogènes/génétique , Récepteur alpha des oestrogènes/immunologie , Femelle , Analyse de profil d'expression de gènes , Humains , Immunité innée , Facteurs de régulation d'interféron/génétique , Facteurs de régulation d'interféron/immunologie , Études longitudinales , Lymphocytes TIL/immunologie , Lymphocytes TIL/anatomopathologie , Maladie résiduelle , Récepteur ErbB-2/génétique , Récepteur ErbB-2/immunologie , Trastuzumab/usage thérapeutique , Résultat thérapeutique , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie
7.
J Immunother Cancer ; 8(2)2020 09.
Article de Anglais | MEDLINE | ID: mdl-32900860

RÉSUMÉ

BACKGROUND: OX40 (CD134) is a costimulatory molecule of the tumor necrosis factor receptor superfamily that is currently being investigated as a target for cancer immunotherapy. However, despite promising results in murine tumor models, the clinical efficacy of agonistic αOX40 antibodies in the treatment of patients with cancer has fallen short of the high expectation in earlier-stage trials. METHODS: Using lymphocytes from resected tumor, tumor-free (TF) tissue and peripheral blood mononuclear cells (PBMC) of 96 patients with hepatocellular and colorectal cancers, we determined OX40 expression and the in vitro T-cell agonistic activity of OX40-targeting compounds. RNA-Seq was used to evaluate OX40-mediated transcriptional changes in CD4+ and CD8+ human tumor-infiltrating lymphocytes (TILs). RESULTS: Here, we show that OX40 was overexpressed on tumor-infiltrating CD4+ T cells compared with blood and TF tissue-derived T cells. In contrast to a clinical candidate αOX40 antibody, treatment with an Fc-engineered αOX40 antibody (αOX40_v12) with selectively enhanced FcγRIIB affinity, stimulated in vitro CD4+ and CD8+ TIL expansion, as well as cytokine and chemokine secretions. The activity of αOX40_v12 was dependent on FcγRIIB engagement and intrinsic CD3/CD28 signals. The transcriptional landscape of CD4+ and CD8+ TILs shifted toward a prosurvival, inflammatory and chemotactic profile on treatment with αOX40_v12. CONCLUSIONS: OX40 is overexpressed on CD4+ TILs and thus represents a promising target for immunotherapy. Targeting OX40 with currently used agonistic antibodies may be inefficient due to lack of OX40 multimerization. Thus, Fc engineering is a powerful tool in enhancing the agonistic activity of αOX40 antibody and may shape the future design of antibody-mediated αOX40 immunotherapy.


Sujet(s)
Immunothérapie/méthodes , Lymphocytes TIL/immunologie , Récepteur au OX40/immunologie , Lymphocytes T/immunologie , Animaux , Modèles animaux de maladie humaine , Femelle , Humains , Mâle , Souris
8.
Nat Med ; 26(11): 1733-1741, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32895571

RÉSUMÉ

We report on molecular analyses of baseline tumor samples from the phase 3 JAVELIN Renal 101 trial (n = 886; NCT02684006 ), which demonstrated significantly prolonged progression-free survival (PFS) with first-line avelumab + axitinib versus sunitinib in advanced renal cell carcinoma (aRCC). We found that neither expression of the commonly assessed biomarker programmed cell death ligand 1 (PD-L1) nor tumor mutational burden differentiated PFS in either study arm. Similarly, the presence of FcɣR single nucleotide polymorphisms was unimpactful. We identified important biological features associated with differential PFS between the treatment arms, including new immunomodulatory and angiogenesis gene expression signatures (GESs), previously undescribed mutational profiles and their corresponding GESs, and several HLA types. These findings provide insight into the determinants of response to combined PD-1/PD-L1 and angiogenic pathway inhibition and may aid in the development of strategies for improved patient care in aRCC.


Sujet(s)
Anticorps monoclonaux humanisés/administration et posologie , Axitinib/administration et posologie , Marqueurs biologiques tumoraux/génétique , Néphrocarcinome/traitement médicamenteux , Sunitinib/administration et posologie , Adolescent , Adulte , Sujet âgé , Anticorps monoclonaux/administration et posologie , Anticorps monoclonaux/effets indésirables , Anticorps monoclonaux humanisés/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Axitinib/effets indésirables , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Mâle , Adulte d'âge moyen , Survie sans progression , Sunitinib/effets indésirables , Transcriptome , Jeune adulte
9.
Clin Cancer Res ; 26(11): 2524-2534, 2020 06 01.
Article de Anglais | MEDLINE | ID: mdl-32144134

RÉSUMÉ

PURPOSE: In this phase I study (NCT01307267), we evaluated safety, pharmacokinetics, clinical activity, and pharmacodynamics of treatment with utomilumab plus rituximab in patients with relapsed/refractory follicular lymphoma (FL) and other CD20+ non-Hodgkin lymphomas (NHL). PATIENTS AND METHODS: Primary objectives were to assess treatment safety and tolerability for estimating the MTD, using a modified time-to-event continual reassessment method, and selecting the recommended phase II dose (RP2D). RESULTS: Sixty-seven patients received utomilumab (0.03-10.0 mg/kg every 4 weeks) and rituximab (375 mg/m2 weekly) in the dose-escalation groups or utomilumab (1.2 mg/kg every 4 weeks) plus rituximab in the dose-expansion cohort. No patient experienced dose-limiting toxicity. The MTD for utomilumab in combination with rituximab was not reached and estimated to be ≥10 mg/kg every 4 weeks. The majority of the utomilumab treatment-related adverse events (AE) were grade 1 to 2; the most common AE was fatigue (16.4%). The pharmacokinetics of utomilumab in combination with rituximab was linear in the 0.03 to 10 mg/kg dose range. A low incidence (1.5%) of treatment-induced antidrug antibodies against utomilumab was observed. The objective response rate was 21.2% (95% CI, 12.1%-33.0%) in all patients with NHL, including four complete and 10 partial responses. Analysis of paired biopsies from a relapsed/refractory FL patient with complete response showed increased T-cell infiltration and cytotoxic activity in tumors. Biomarker correlations with outcomes suggested that clinical benefit may be contingent on patient immune function. CONCLUSIONS: Utomilumab in combination with rituximab demonstrated clinical activity and a favorable safety profile in patients with CD20+ NHLs.


Sujet(s)
Adénocarcinome folliculaire/traitement médicamenteux , Antigènes CD20/métabolisme , Protocoles de polychimiothérapie antinéoplasique/pharmacocinétique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lymphome malin non hodgkinien/traitement médicamenteux , Antigènes CD137/agonistes , Adénocarcinome folliculaire/immunologie , Adénocarcinome folliculaire/anatomopathologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Anticorps monoclonaux humanisés/administration et posologie , Femelle , Études de suivi , Humains , Immunoglobuline G/administration et posologie , Lymphome malin non hodgkinien/immunologie , Lymphome malin non hodgkinien/anatomopathologie , Mâle , Dose maximale tolérée , Adulte d'âge moyen , Pronostic , Rituximab/administration et posologie , Distribution tissulaire
10.
Cell Rep ; 30(6): 1935-1950.e8, 2020 02 11.
Article de Anglais | MEDLINE | ID: mdl-32049022

RÉSUMÉ

Alternative splicing is well understood to enhance proteome diversity as cells respond to stimuli. However, mechanistic understanding for how the spliceosome processes precursor messenger RNA (mRNA) transcripts to achieve template diversification is incomplete. We use recently developed enzymatic inhibitors of protein arginine methyltransferase 5 (PRMT5) and human naive T lymphocyte activation as a model system to uncover a precise set of mRNA transcripts that require symmetric arginine dimethylation. This methylation-dependent splicing selectivity is associated with a limited set of signaling pathways that are affected when PRMT5 is inhibited. Specifically, we identify a conserved role for symmetric arginine dimethylation in the induction of antiviral type I and type III interferon signaling following T cell receptor and pattern recognition receptor stimulation in human T lymphocytes and undifferentiated human THP-1 monocytes. Altogether, these findings reveal a mechanism by which cells may be enabled to precisely modulate transcript heterogeneity to orchestrate specific functional outcomes.


Sujet(s)
Épissage alternatif/génétique , Arginine/métabolisme , Interférons/métabolisme , Épissage des ARN/génétique , Humains , Transduction du signal
11.
Clin Cancer Res ; 21(6): 1487-96, 2015 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-25564152

RÉSUMÉ

PURPOSE: To identify and characterize novel, activating mutations in Notch receptors in breast cancer and to determine response to the gamma secretase inhibitor (GSI) PF-03084014. EXPERIMENTAL DESIGN: We used several computational approaches, including novel algorithms, to analyze next-generation sequencing data and related omic datasets from The Cancer Genome Atlas (TCGA) breast cancer cohort. Patient-derived xenograft (PDX) models were sequenced, and Notch-mutant models were treated with PF-03084014. Gene-expression and functional analyses were performed to study the mechanism of activation through mutation and inhibition by PF-03084014. RESULTS: We identified mutations within and upstream of the PEST domains of NOTCH1, NOTCH2, and NOTCH3 in the TCGA dataset. Mutations occurred via several genetic mechanisms and compromised the function of the PEST domain, a negative regulatory domain commonly mutated in other cancers. Focal amplifications of NOTCH2 and NOTCH3 were also observed, as were heterodimerization or extracellular domain mutations at lower incidence. Mutations and amplifications often activated the Notch pathway as evidenced by increased expression of canonical Notch target genes, and functional mutations were significantly enriched in the triple-negative breast cancer subtype (TNBC). PDX models were also identified that harbored PEST domain mutations, and these models were highly sensitive to PF-03084014. CONCLUSIONS: This work suggests that Notch-altered breast cancer constitutes a bona fide oncogenic driver segment with the most common alteration being PEST domain mutations present in multiple Notch receptors. Importantly, functional studies suggest that this newly identified class can be targeted with Notch inhibitors, including GSIs.


Sujet(s)
Amyloid precursor protein secretases/antagonistes et inhibiteurs , Récepteur Notch1/génétique , Récepteur Notch2/génétique , Récepteurs Notch/génétique , Tumeurs du sein triple-négatives/traitement médicamenteux , Algorithmes , Animaux , Séquence nucléotidique , Lignée cellulaire tumorale , Prolifération cellulaire , Biologie informatique/méthodes , Variations de nombre de copies de segment d'ADN/génétique , Femelle , Dosage génique/génétique , Humains , Souris , Souris SCID , Structure tertiaire des protéines/génétique , Récepteur Notch3 , Analyse de séquence d'ADN , Transduction du signal/génétique , 1,2,3,4-Tétrahydro-naphtalènes/pharmacologie , Tumeurs du sein triple-négatives/génétique , Valine/analogues et dérivés , Valine/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
12.
Pac Symp Biocomput ; : 10-9, 2015.
Article de Anglais | MEDLINE | ID: mdl-25592564

RÉSUMÉ

The Cell Index Database, (CELLX) (http://cellx.sourceforge.net) provides a computational framework for integrating expression, copy number variation, mutation, compound activity, and meta data from cancer cells. CELLX provides the computational biologist a quick way to perform routine analyses as well as the means to rapidly integrate data for offline analysis. Data is accessible through a web interface which utilizes R to generate plots and perform clustering, correlations, and statistical tests for associations within and between data types for ~20,000 samples from TCGA, CCLE, Sanger, GSK, GEO, GTEx, and other public sources. We show how CELLX supports precision oncology through indications discovery, biomarker evaluation, and cell line screening analysis.


Sujet(s)
Bases de données génétiques , Tumeurs/génétique , Logiciel , Marqueurs biologiques tumoraux/génétique , Lignée cellulaire tumorale , Biologie informatique , Humains , Internet , Tumeurs/thérapie , Médecine de précision
13.
Genomics ; 103(2-3): 189-203, 2014.
Article de Anglais | MEDLINE | ID: mdl-24462510

RÉSUMÉ

Elucidating the molecular basis of hepatocellular carcinoma (HCC) is crucial to developing targeted diagnostics and therapies for this deadly disease. The landscape of somatic genomic rearrangements (GRs), which can lead to oncogenic gene fusions, remains poorly characterized in HCC. We have predicted 4314 GRs including large-scale insertions, deletions, inversions and translocations based on the whole-genome sequencing data for 88 primary HCC tumor/non-tumor tissues. We identified chromothripsis in 5 HCC genomes (5.7%) recurrently affecting chromosomal arms 1q and 8q. Albumin (ALB) was found to harbor GRs, deactivating mutations and deletions in 10% of cohort. Integrative analysis identified a pattern of paired intra-chromosomal translocations flanking focal amplifications and asymmetrical patterns of copy number variation flanking breakpoints of translocations. Furthermore, we predicted 260 gene fusions which frequently result in aberrant over-expression of the 3' genes in tumors and validated 18 gene fusions, including recurrent fusion (2/88) of ABCB11 and LRP2.


Sujet(s)
Carcinome hépatocellulaire/génétique , Réarrangement des gènes , Génome humain , Tumeurs du foie/génétique , Translocation génétique , Chromosomes humains de la paire 1/génétique , Chromosomes humains de la paire 8/génétique , Études de cohortes , Femelle , Étude d'association pangénomique/méthodes , Humains , Mâle
14.
Mol Cancer Ther ; 12(12): 2929-39, 2013 Dec.
Article de Anglais | MEDLINE | ID: mdl-24107449

RÉSUMÉ

Figitumumab (CP-751,871), a potent and fully human monoclonal anti-insulin-like growth factor 1 receptor (IGF1R) antibody, has been investigated in clinical trials of several solid tumors. To identify biomarkers of sensitivity and resistance to figitumumab, its in vitro antiproliferative activity was analyzed in a panel of 93 cancer cell lines by combining in vitro screens with extensive molecular profiling of genomic aberrations. Overall response was bimodal and the majority of cell lines were resistant to figitumumab. Nine of 15 sensitive cell lines were derived from colon cancers. Correlations between genomic characteristics of cancer cell lines with figitumumab antiproliferative activity revealed that components of the IGF pathway, including IRS2 (insulin receptor substrate 2) and IGFBP5 (IGF-binding protein 5), played a pivotal role in determining the sensitivity of tumors to single-agent figitumumab. Tissue-specific differences among the top predictive genes highlight the need for tumor-specific patient selection strategies. For the first time, we report that alteration or expression of the MYB oncogene is associated with sensitivity to IGF1R inhibitors. MYB is dysregulated in hematologic and epithelial tumors, and IGF1R inhibition may represent a novel therapeutic opportunity. Although growth inhibitory activity with single-agent figitumumab was relatively rare, nine combinations comprising figitumumab plus chemotherapeutic agents or other targeted agents exhibited properties of synergy. Inhibitors of the ERBB family were frequently synergistic and potential biomarkers of drug synergy were identified. Several biomarkers of antiproliferative activity of figitumumab both alone and in combination with other therapies may inform the design of clinical trials evaluating IGF1R inhibitors.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Antinéoplasiques/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Récepteur IGF de type 1/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Analyse de regroupements , Variations de nombre de copies de segment d'ADN , Synergie des médicaments , Analyse de profil d'expression de gènes , Humains , Concentration inhibitrice 50 , Mutation , Locus de caractère quantitatif , Récepteur IGF de type 1/métabolisme , Transduction du signal
16.
PLoS One ; 7(6): e39653, 2012.
Article de Anglais | MEDLINE | ID: mdl-22745804

RÉSUMÉ

Targeting cancers with amplified or abnormally activated c-Met (hepatocyte growth factor receptor) may have therapeutic benefit based on nonclinical and emerging clinical findings. However, the eventual emergence of drug resistant tumors motivates the pre-emptive identification of potential mechanisms of clinical resistance. We rendered a MET amplified gastric cancer cell line, GTL16, resistant to c-Met inhibition with prolonged exposure to a c-Met inhibitor, PF-04217903 (METi). Characterization of surviving cells identified an amplified chromosomal rearrangement between 7q32 and 7q34 which overexpresses a constitutively active SND1-BRAF fusion protein. In the resistant clones, hyperactivation of the downstream MAPK pathway via SND1-BRAF conferred resistance to c-Met receptor tyrosine kinase inhibition. Combination treatment with METi and a RAF inhibitor, PF-04880594 (RAFi) inhibited ERK activation and circumvented resistance to either single agent. Alternatively, treatment with a MEK inhibitor, PD-0325901 (MEKi) alone effectively blocked ERK phosphorylation and inhibited cell growth. Our results suggest that combination of a c-Met tyrosine kinase inhibitor with a BRAF or a MEK inhibitor may be effective in treating resistant tumors that use activated BRAF to escape suppression of c-Met signaling.


Sujet(s)
Mitogen-Activated Protein Kinases/métabolisme , Protéines nucléaires/métabolisme , Protéines proto-oncogènes B-raf/métabolisme , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Pyrazines/pharmacologie , Protéines de fusion recombinantes/métabolisme , Triazoles/pharmacologie , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques , Endonucleases , Humains , Protéines nucléaires/génétique , Protéines proto-oncogènes B-raf/génétique , Protéines de fusion recombinantes/génétique
17.
Mol Cancer Ther ; 11(3): 710-9, 2012 Mar.
Article de Anglais | MEDLINE | ID: mdl-22222631

RÉSUMÉ

PF-03814735 is a novel, reversible inhibitor of Aurora kinases A and B that finished a phase I clinical trial for the treatment of advanced solid tumors. To find predictive biomarkers of drug sensitivity, we screened a diverse panel of 87 cancer cell lines for growth inhibition upon PF-03814735 treatment. Small cell lung cancer (SCLC) and, to a lesser extent, colon cancer lines were very sensitive to PF-03814735. The status of the Myc gene family and retinoblastoma pathway members significantly correlated with the efficacy of PF-03814735. Whereas RB1 inactivation, intact CDKN2A/p16, and normal CCND1/Cyclin D1 status are hallmarks of SCLC, activation or amplification of any of the three Myc genes (MYC, MYCL1, and MYCN) clearly differentiated cell line sensitivity within the SCLC panel. By contrast, we found that expression of Aurora A and B were weak predictors of response. We observed a decrease in histone H3 phosphorylation and polyploidization of sensitive lines, consistent with the phenotype of Aurora B inhibition. In vivo experiments with two SCLC xenograft models confirmed the sensitivity of Myc gene-driven models to PF-03814735 and a possible schedule dependence of MYC/c-Myc-driven tumors. Altogether our results suggest that SCLC and other malignancies driven by the Myc family genes may be suitable indications for treatment by Aurora B kinase inhibitors.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Composés hétérocycliques 3 noyaux/pharmacologie , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Pyrimidines/pharmacologie , Animaux , Antinéoplasiques/pharmacologie , Aurora kinase A , Aurora kinase B , Aurora kinases , Marqueurs biologiques tumoraux/métabolisme , Technique de Western , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Analyse de profil d'expression de gènes , Génomique/méthodes , Histone/métabolisme , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Souris , Souris nude , Phosphorylation/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes c-myc/métabolisme , Interférence par ARN , RT-PCR , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Carcinome pulmonaire à petites cellules/génétique , Carcinome pulmonaire à petites cellules/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
18.
Mol Cancer Ther ; 11(2): 439-51, 2012 Feb.
Article de Anglais | MEDLINE | ID: mdl-22135232

RÉSUMÉ

Recently, HER2-directed treatment, such as trastuzumab, has shown clinical benefit in HER2-amplified gastric cancer. On the basis of recent studies about epidermal growth factor receptor (EGFR) or HER2-targeting agents (including gefitinib, lapatinib, and trastuzumab) in gastric cancer, the potent effects of pan-HER inhibitors targeting the HER family are anticipated. In this study, we evaluated the activity and mechanisms of PF00299804, an irreversible pan-HER inhibitor, in gastric cancer in vitro and in vivo models. PF00299804 showed significant growth-inhibitory effects in HER2-amplified gastric cancer cells (SNU216, N87), and it had lower 50% inhibitory concentration values compared with other EGFR tyrosine kinase inhibitors, including gefitinib, lapatinib, BIBW-2992, and CI-1033. PF00299804 induced apoptosis and G(1) arrest and inhibited phosphorylation of receptors in the HER family and downstream signaling pathways including STAT3, AKT, and extracellular signal-regulated kinases (ERK) in HER2-amplified gastric cancer cells. PF00299804 also blocked EGFR/HER2, HER2/HER3, and HER3/HER4 heterodimer formation as well as the association of HER3 with p85α in SNU216 cells. The combination of PF00299804 with clinically relevant chemotherapeutic agents or molecular-targeted agents including trastuzumab (an anti-HER2 monoclonal antibody), CP751871 (an IGF1R inhibitor), PD0325901 (an ERK1/2 inhibitor), and PF04691502 (a PI3K/mTOR inhibitor) produced synergistic effects. These findings indicate that PF00299804 can be used as a targeted therapy for the treatment of HER2-amplified gastric cancer through inhibition of HER family heterodimer formation and may augment antitumor efficacy of chemotherapeutic and/or molecular-targeted agents.


Sujet(s)
Antinéoplasiques/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Quinazolinones/pharmacologie , Tumeurs de l'estomac/traitement médicamenteux , Animaux , Anticorps monoclonaux humanisés/administration et posologie , Anticorps monoclonaux humanisés/pharmacologie , Antinéoplasiques/administration et posologie , Antinéoplasiques/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Technique de Western , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Évaluation préclinique de médicament , Synergie des médicaments , Femelle , Phase G1/effets des médicaments et des substances chimiques , Humains , Souris , Souris SCID , Structure moléculaire , Inhibiteurs de protéines kinases/administration et posologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Quinazolinones/administration et posologie , Quinazolinones/composition chimique , Récepteur ErbB-2/antagonistes et inhibiteurs , Récepteur ErbB-2/métabolisme , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/anatomopathologie , Facteurs temps , Trastuzumab , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
19.
Cell Res ; 21(10): 1393-409, 2011 Oct.
Article de Anglais | MEDLINE | ID: mdl-21876557

RÉSUMÉ

Pluripotency, the ability of a cell to differentiate and give rise to all embryonic lineages, defines a small number of mammalian cell types such as embryonic stem (ES) cells. While it has been generally held that pluripotency is the product of a transcriptional regulatory network that activates and maintains the expression of key stem cell genes, accumulating evidence is pointing to a critical role for epigenetic processes in establishing and safeguarding the pluripotency of ES cells, as well as maintaining the identity of differentiated cell types. In order to better understand the role of epigenetic mechanisms in pluripotency, we have examined the dynamics of chromatin modifications genome-wide in human ES cells (hESCs) undergoing differentiation into a mesendodermal lineage. We found that chromatin modifications at promoters remain largely invariant during differentiation, except at a small number of promoters where a dynamic switch between acetylation and methylation at H3K27 marks the transition between activation and silencing of gene expression, suggesting a hierarchy in cell fate commitment over most differentially expressed genes. We also mapped over 50 000 potential enhancers, and observed much greater dynamics in chromatin modifications, especially H3K4me1 and H3K27ac, which correlate with expression of their potential target genes. Further analysis of these enhancers revealed potentially key transcriptional regulators of pluripotency and a chromatin signature indicative of a poised state that may confer developmental competence in hESCs. Our results provide new evidence supporting the role of chromatin modifications in defining enhancers and pluripotency.


Sujet(s)
Différenciation cellulaire/physiologie , Cellules souches embryonnaires/métabolisme , Épigenèse génétique/physiologie , Cellules souches pluripotentes/métabolisme , Transcription génétique/physiologie , Lignée cellulaire , Lignage cellulaire/physiologie , Chromatine/génétique , Chromatine/métabolisme , Cellules souches embryonnaires/cytologie , Éléments activateurs (génétique)/physiologie , Étude d'association pangénomique , Humains , Cellules souches pluripotentes/cytologie
20.
Nature ; 459(7243): 108-12, 2009 May 07.
Article de Anglais | MEDLINE | ID: mdl-19295514

RÉSUMÉ

The human body is composed of diverse cell types with distinct functions. Although it is known that lineage specification depends on cell-specific gene expression, which in turn is driven by promoters, enhancers, insulators and other cis-regulatory DNA sequences for each gene, the relative roles of these regulatory elements in this process are not clear. We have previously developed a chromatin-immunoprecipitation-based microarray method (ChIP-chip) to locate promoters, enhancers and insulators in the human genome. Here we use the same approach to identify these elements in multiple cell types and investigate their roles in cell-type-specific gene expression. We observed that the chromatin state at promoters and CTCF-binding at insulators is largely invariant across diverse cell types. In contrast, enhancers are marked with highly cell-type-specific histone modification patterns, strongly correlate to cell-type-specific gene expression programs on a global scale, and are functionally active in a cell-type-specific manner. Our results define over 55,000 potential transcriptional enhancers in the human genome, significantly expanding the current catalogue of human enhancers and highlighting the role of these elements in cell-type-specific gene expression.


Sujet(s)
Phénomènes physiologiques cellulaires , Régulation de l'expression des gènes , Histone/métabolisme , Facteurs de transcription/génétique , Sites de fixation , Lignée cellulaire , Chromatine/génétique , Génome humain/génétique , Cellules HeLa , Humains , Cellules K562 , Régions promotrices (génétique)/génétique , Facteurs de transcription/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...