Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 24
Filtrer
1.
J Biomed Inform ; 142: 104367, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37105509

RÉSUMÉ

Cytokine release syndrome (CRS), also known as cytokine storm, is one of the most consequential adverse effects of chimeric antigen receptor therapies that have shown otherwise promising results in cancer treatment. When emerging, CRS could be identified by the analysis of specific cytokine and chemokine profiles that tend to exhibit similarities across patients. In this paper, we exploit these similarities using machine learning algorithms and set out to pioneer a meta-review informed method for the identification of CRS based on specific cytokine peak concentrations and evidence from previous clinical studies. To this end we also address a widespread challenge of the applicability of machine learning in general: reduced training data availability. We do so by augmenting available (but often insufficient) patient cytokine concentrations with statistical knowledge extracted from domain literature. We argue that such methods could support clinicians in analyzing suspect cytokine profiles by matching them against the said CRS knowledge from past clinical studies, with the ultimate aim of swift CRS diagnosis. We evaluate our proposed methods under several design choices, achieving performance of more than 90% in terms of CRS identification accuracy, and showing that many of our choices outperform a purely data-driven alternative. During evaluation with real-world CRS clinical data, we emphasize the potential of our proposed method of producing interpretable results, in addition to being effective in identifying the onset of cytokine storm.


Sujet(s)
Récepteurs chimériques pour l'antigène , Humains , Thérapie cellulaire et tissulaire , Syndrome de libération de cytokines/diagnostic , Cytokines , Immunothérapie adoptive/méthodes
2.
Clin Cancer Res ; 28(10): 1999-2019, 2022 05 13.
Article de Anglais | MEDLINE | ID: mdl-35091446

RÉSUMÉ

Poorly differentiated neuroendocrine carcinomas (PD-NEC) are rare cancers garnering interest as they become more commonly encountered in the clinic. This is due to improved diagnostic methods and the increasingly observed phenomenon of "NE lineage plasticity," whereby nonneuroendocrine (non-NE) epithelial cancers transition to aggressive NE phenotypes after targeted treatment. Effective treatment options for patients with PD-NEC are challenging for several reasons. This includes a lack of targetable, recurrent molecular drivers, a paucity of patient-relevant preclinical models to study biology and test novel therapeutics, and the absence of validated biomarkers to guide clinical management. Although advances have been made pertaining to molecular subtyping of small cell lung cancer (SCLC), a PD-NEC of lung origin, extrapulmonary (EP)-PD-NECs remain understudied. This review will address emerging SCLC-like, same-organ non-NE cancer-like and tumor-type-agnostic biological vulnerabilities of EP-PD-NECs, with the potential for therapeutic exploitation. The hypotheses surrounding the origin of these cancers and how "NE lineage plasticity" can be leveraged for therapeutic purposes are discussed. SCLC is herein proposed as a paradigm for supporting progress toward precision medicine in EP-PD-NECs. The aim of this review is to provide a thorough portrait of the current knowledge of EP-PD-NEC biology, with a view to informing new avenues for research and future therapeutic opportunities in these cancers of unmet need.


Sujet(s)
Carcinome neuroendocrine , Tumeurs du poumon , Tumeurs neuroendocrines , Carcinome pulmonaire à petites cellules , Marqueurs biologiques tumoraux/usage thérapeutique , Carcinome neuroendocrine/diagnostic , Carcinome neuroendocrine/traitement médicamenteux , Carcinome neuroendocrine/génétique , Humains , Nouveau-né , Poumon/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs neuroendocrines/traitement médicamenteux , Tumeurs neuroendocrines/anatomopathologie , Carcinome pulmonaire à petites cellules/anatomopathologie
3.
Mol Oncol ; 16(10): 1969-1985, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-34866317

RÉSUMÉ

Nearly all estrogen receptor (ER)-positive (POS) metastatic breast cancers become refractory to endocrine (ET) and other therapies, leading to lethal disease presumably due to evolving genomic alterations. Timely monitoring of the molecular events associated with response/progression by serial tissue biopsies is logistically difficult. Use of liquid biopsies, including circulating tumor cells (CTC) and circulating tumor DNA (ctDNA), might provide highly informative, yet easily obtainable, evidence for better precision oncology care. Although ctDNA profiling has been well investigated, the CTC precision oncology genomic landscape and the advantages it may offer over ctDNA in ER-POS breast cancer remain largely unexplored. Whole-blood (WB) specimens were collected at serial time points from patients with advanced ER-POS/HER2-negative (NEG) advanced breast cancer in a phase I trial of AZD9496, an oral selective ER degrader (SERD) ET. Individual CTC were isolated from WB using tandem CellSearch® /DEPArray™ technologies and genomically profiled by targeted single-cell DNA next-generation sequencing (scNGS). High-quality CTC (n = 123) from 12 patients profiled by scNGS showed 100% concordance with ctDNA detection of driver estrogen receptor α (ESR1) mutations. We developed a novel CTC-based framework for precision medicine actionability reporting (MI-CTCseq) that incorporates novel features, such as clonal predominance and zygosity of targetable alterations, both unambiguously identifiable in CTC compared to ctDNA. Thus, we nominated opportunities for targeted therapies in 73% of patients, directed at alterations in phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), fibroblast growth factor receptor 2 (FGFR2), and KIT proto-oncogene, receptor tyrosine kinase (KIT). Intrapatient, inter-CTC genomic heterogeneity was observed, at times between time points, in subclonal alterations. Our analysis suggests that serial monitoring of the CTC genome is feasible and should enable real-time tracking of tumor evolution during progression, permitting more combination precision medicine interventions.


Sujet(s)
Tumeurs du sein , ADN tumoral circulant , Cellules tumorales circulantes , Marqueurs biologiques tumoraux/génétique , Tumeurs du sein/anatomopathologie , ADN tumoral circulant/génétique , Antagonistes des oestrogènes , Études de faisabilité , Femelle , Génomique , Humains , Mutation/génétique , Cellules tumorales circulantes/anatomopathologie , Médecine de précision
4.
Br J Cancer ; 126(5): 693-705, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34663949

RÉSUMÉ

Biliary tract cancers, including intra- and extra-hepatic cholangiocarcinoma as well as gallbladder cancer, are associated with poor prognosis and the majority of patients present with advanced-stage, non-resectable disease at diagnosis. Biliary tract cancer may develop through an accumulation of genetic and epigenetic alterations and can be influenced by microbial exposure. Furthermore, the liver and biliary tract are exposed to the gastrointestinal microbiome through the gut-liver axis. The availability of next-generation sequencing technology has led to an increase in studies investigating the relationship between microbiota and human disease. In particular, the interplay between the microbiome, the tumour micro-environment and response to systemic therapy is a prospering area of interest. Given the poor outcomes for patients with biliary tract cancer, this emerging field of research, through which new biomarkers may be identified, offers potential as a tool for early diagnosis, prognostication or even as a future therapeutic target. This review summarises the available evidence on the microbiome environment in patients with biliary tract cancer, including a discussion around confounding factors, implications for therapy and proposed future directions.


Sujet(s)
Bactéries/classification , Tumeurs des voies biliaires/microbiologie , Tumeurs des voies biliaires/traitement médicamenteux , Tumeurs des voies biliaires/génétique , Épigenèse génétique , Microbiome gastro-intestinal , Séquençage nucléotidique à haut débit , Humains , Pronostic , Microenvironnement tumoral
5.
Clin Cancer Res ; 27(4): 1119-1130, 2021 02 15.
Article de Anglais | MEDLINE | ID: mdl-33257424

RÉSUMÉ

PURPOSE: At diagnosis, colorectal cancer presents with synchronous peritoneal metastasis in up to 10% of patients. The peritoneum is poorly characterized with respect to its superspecialized microenvironment. Our aim was to describe the differences between peritoneal metastases and their matched primary tumors excised simultaneously at the time of surgery. Also, we tested the hypothesis of these differences being present in primary colorectal tumors and having prognostic capacity. EXPERIMENTAL DESIGN: We report a comprehensive analysis of 30 samples from peritoneal metastasis with their matched colorectal cancer primaries obtained during cytoreductive surgery. We tested and validated the prognostic value of our findings in a pooled series of 660 colorectal cancer primary samples with overall survival (OS) information and 743 samples with disease-free survival (DFS) information from publicly available databases. RESULTS: We identified 20 genes dysregulated in peritoneal metastasis that promote an early increasing role of "stemness" in conjunction with tumor-favorable inflammatory changes. When adjusted for age, gender, and stage, the 20-gene peritoneal signature proved to have prognostic value for both OS [adjusted HR for the high-risk group (vs. low-risk) 2.32 (95% confidence interval, CI, 1.69-3.19; P < 0.0001)] and for DFS [adjusted HR 2.08 (95% CI, 1.50-2.91; P < 0.0001)]. CONCLUSIONS: Our findings indicated that the activation of "stemness" pathways and adaptation to the peritoneal-specific environment are key to early stages of peritoneal carcinomatosis. The in silico analysis suggested that this 20-gene peritoneal signature may hold prognostic information with potential for development of new precision medicine strategies in this setting.


Sujet(s)
Tumeurs colorectales/anatomopathologie , Récidive tumorale locale/épidémiologie , Cellules souches tumorales/anatomopathologie , Cavité péritonéale/anatomopathologie , Tumeurs du péritoine/immunologie , Adulte , Sujet âgé , Tumeurs colorectales/immunologie , Tumeurs colorectales/mortalité , Tumeurs colorectales/thérapie , Interventions chirurgicales de cytoréduction , Survie sans rechute , Femelle , Études de suivi , Humains , Chimiothérapie hyperthermique intrapéritonéale , Mâle , Adulte d'âge moyen , Récidive tumorale locale/immunologie , Récidive tumorale locale/anatomopathologie , Tumeurs du péritoine/mortalité , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/thérapie , Appréciation des risques/méthodes , Microenvironnement tumoral/immunologie
6.
Cancer Cell ; 37(4): 485-495, 2020 04 13.
Article de Anglais | MEDLINE | ID: mdl-32289272

RÉSUMÉ

Predictive biomarkers aid selection of personalized therapy targeted to molecular alterations within an individual's tumor. Patients' responses to targeted therapies are commonly followed by treatment resistance. Here, we survey liquid biopsies as alternatives to tumor biopsies to assess predictive and therapy response biomarkers. We examine the potential of liquid biopsies to meet the challenges of minimal residual disease monitoring after curative intent treatment for earlier detection of disease recurrence. We focus on blood, the most commonly collected minimally invasive clinical sample, and on the two most widely studied assays, circulating tumor DNA and circulating tumor cells.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Marqueurs biologiques tumoraux/génétique , ADN tumoral/analyse , Résistance aux médicaments antinéoplasiques/génétique , Biopsie liquide/méthodes , Tumeurs/anatomopathologie , Cellules tumorales circulantes/anatomopathologie , ADN tumoral/génétique , Humains , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Médecine de précision
7.
Br J Cancer ; 120(2): 141-153, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30580378

RÉSUMÉ

Cancers of Unknown Primary (CUP) comprise a heterogeneous clinical entity of confirmed metastatic cancer where the primary site of origin is undetectable. It has a poor prognosis with limited treatment options. CUP is historically under-researched; however, understanding its biology has the potential to not only improve treatment and survival by implementation of biomarkers for patient management, but also to greatly contribute to our understanding of carcinogenesis and metastasis across all cancer types. Here we review the current advances in CUP research and explore the debated hypotheses underlying its biology. The evolution of molecular profiling and tissue-of-origin classifiers have the potential to transform the diagnosis, classification and therapeutic management of patients with CUP but robust evidence to support widespread use is lacking. Precision medicine has transformed treatment strategy in known tumour types; in CUP, however, there remains a clinical need for a better understanding of molecular characteristics to establish the potential role of novel or existing therapeutics. The emergence of liquid biopsies as a source of predictive and prognostic biomarkers within known tumour types is gaining rapid ground and this review explores the potential utility of liquid biopsies in CUP.


Sujet(s)
Marqueurs biologiques tumoraux/sang , Carcinomes/génétique , Métastases d'origine inconnue/génétique , Pronostic , Carcinomes/sang , Régulation de l'expression des gènes tumoraux , Humains , Métastases d'origine inconnue/sang , Médecine de précision
8.
Lancet Oncol ; 19(9): e470-e481, 2018 09.
Article de Anglais | MEDLINE | ID: mdl-30191851

RÉSUMÉ

Small-cell lung cancer (SCLC) is an aggressive tumour that seeds metastases early with dismal outcomes. As expected from a disease that is closely associated with smoking, mutation burden in SCLC is high. Intratumoral and intertumoral heterogeneity is a substantial obstacle to successful treatment and the SCLC genomic landscape reveals few targets that are readily druggable. Chemotherapy elicits responses in most patients with SCLC, but their effects are short lived. Multiple clinical trials have been unsuccessful in showing positive survival outcomes and biomarkers to select patients and monitor responses to novel targeted treatments have been lacking, not least because acquisition of tumour biopsies, especially during relapse after chemotherapy, is a substantial challenge. Liquid biopsies via blood sampling in SCLC, notably circulating tumour cells and circulating free tumour DNA can be readily and repeatedly accessed, and are beginning to yield promising data to inform SCLC biology and patient treatment. Primary cell cultures and preclinical mouse models can also be derived from the relatively plentiful SCLC circulating tumour cells providing a tractable platform for SCLC translational research and drug development.


Sujet(s)
ADN tumoral circulant/sang , Biopsie liquide , Tumeurs du poumon/diagnostic , Cellules tumorales circulantes/composition chimique , Cellules tumorales circulantes/anatomopathologie , Carcinome pulmonaire à petites cellules/diagnostic , Animaux , ADN tumoral circulant/génétique , Prise de décision clinique , Humains , Tumeurs du poumon/sang , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Thérapie moléculaire ciblée , Stadification tumorale , Sélection de patients , Médecine de précision , Valeur prédictive des tests , Carcinome pulmonaire à petites cellules/sang , Carcinome pulmonaire à petites cellules/génétique , Carcinome pulmonaire à petites cellules/secondaire
9.
J Clin Oncol ; 36(12): 1232-1239, 2018 04 20.
Article de Anglais | MEDLINE | ID: mdl-29528792

RÉSUMÉ

Purpose Uveal melanoma is the most common primary intraocular malignancy in adults with no effective systemic treatment option in the metastatic setting. Selumetinib (AZD6244, ARRY-142886) is an oral, potent, and selective MEK1/2 inhibitor with a short half-life, which demonstrated single-agent activity in patients with metastatic uveal melanoma in a randomized phase II trial. Methods The Selumetinib (AZD6244: ARRY-142886) (Hyd-Sulfate) in Metastatic Uveal Melanoma (SUMIT) study was a phase III, double-blind trial ( ClinicalTrial.gov identifier: NCT01974752) in which patients with metastatic uveal melanoma and no prior systemic therapy were randomly assigned (3:1) to selumetinib (75 mg twice daily) plus dacarbazine (1,000 mg/m2 intravenously on day 1 of every 21-day cycle) or placebo plus dacarbazine. The primary end point was progression-free survival (PFS) by blinded independent central radiologic review. Secondary end points included overall survival and objective response rate. Results A total of 129 patients were randomly assigned to receive selumetinib plus dacarbazine (n = 97) or placebo plus dacarbazine (n = 32). In the selumetinib plus dacarbazine group, 82 patients (85%) experienced a PFS event, compared with 24 (75%) in the placebo plus dacarbazine group (median, 2.8 v 1.8 months); the hazard ratio for PFS was 0.78 (95% CI, 0.48 to 1.27; two-sided P = .32). The objective response rate was 3% with selumetinib plus dacarbazine and 0% with placebo plus dacarbazine (two-sided P = .36). At 37% maturity (n = 48 deaths), analysis of overall survival gave a hazard ratio of 0.75 (95% CI, 0.39 to 1.46; two-sided P = .40). The most frequently reported adverse events (selumetinib plus dacarbazine v placebo plus dacarbazine) were nausea (62% v 19%), rash (57% v 6%), fatigue (44% v 47%), diarrhea (44% v 22%), and peripheral edema (43% v 6%). Conclusion In patients with metastatic uveal melanoma, the combination of selumetinib plus dacarbazine had a tolerable safety profile but did not significantly improve PFS compared with placebo plus dacarbazine.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Mélanome/traitement médicamenteux , Tumeurs de l'uvée/traitement médicamenteux , Adulte , Sujet âgé , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Benzimidazoles/administration et posologie , Benzimidazoles/effets indésirables , Dacarbazine/administration et posologie , Dacarbazine/effets indésirables , Méthode en double aveugle , Femelle , Humains , Mâle , Mélanome/anatomopathologie , Adulte d'âge moyen , Métastase tumorale , Placebo , Survie sans progression , Tumeurs de l'uvée/anatomopathologie
10.
Clin Cancer Res ; 23(18): 5366-5373, 2017 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-28615371

RÉSUMÉ

Purpose: Squamous cell lung cancers (SQCLC) account for 25% of all NSCLCs, yet the prognosis of these patients is poor and treatment options are limited. Amplified FGFR1 is one of the most common oncogenic events in SQCLCs, occurring in approximately 20% of cases. AZD4547 is a potent and selective FGFR1-3 inhibitor with antitumor activity in FGFR1-amplified SQCLC cell lines and patient-derived xenografts.Experimental Design: On the basis of these data, we performed a phase I study of AZD4547 in patients with previously treated stage IV FGFR1-amplified SQCLCs (NCT00979134). FGFR1 amplification (FGFR1:CEP8 ≥ 2) was determined by FISH. The primary endpoint was safety/tolerability. Secondary endpoints included antitumor activity, pharmacokinetics, pharmacodynamics, and molecular analyses.Results: Fifteen FGFR1-amplified patients were treated. The most common related adverse events (AE) were gastrointestinal and dermatologic. Grade ≥3-related AEs occurred in 3 patients (23%). Thirteen patients were evaluable for radiographic response assessment. The overall response rate was 8% (1 PR). Two of 15 patients (13.3%) were progression-free at 12 weeks, and the median overall survival was 4.9 months. Molecular tests, including next-generation sequencing, gene expression analysis, and FGFR1 immunohistochemistry, showed poor correlation between gene amplification and expression, potential genomic modifiers of efficacy, and heterogeneity in 8p11 amplicon.Conclusions: AZD4547 was tolerable at a dosage of 80 mg oral twice a day, with modest antitumor activity. Detailed molecular studies show that these tumors are heterogeneous, with a range of mutational covariates and stark differences in gene expression of the 8p11 amplicon that likely explain the modest efficacy of FGFR inhibition in this disease. Clin Cancer Res; 23(18); 5366-73. ©2017 AACR.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Benzamides/usage thérapeutique , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Pipérazines/usage thérapeutique , Pyrazoles/usage thérapeutique , Sujet âgé , Antinéoplasiques/administration et posologie , Antinéoplasiques/effets indésirables , Antinéoplasiques/pharmacocinétique , Benzamides/administration et posologie , Benzamides/effets indésirables , Benzamides/pharmacocinétique , Carcinome épidermoïde/génétique , Chromosomes humains de la paire 8 , Femelle , Amplification de gène , Analyse de profil d'expression de gènes , Hétérogénéité génétique , Humains , Tumeurs du poumon/génétique , Mâle , Adulte d'âge moyen , Grading des tumeurs , Métastase tumorale , Stadification tumorale , Pipérazines/administration et posologie , Pipérazines/effets indésirables , Pipérazines/pharmacocinétique , Pyrazoles/administration et posologie , Pyrazoles/effets indésirables , Pyrazoles/pharmacocinétique , Récepteur FGFR1/génétique , Récepteur FGFR1/métabolisme , Analyse de séquence d'ADN , Résultat thérapeutique
11.
JAMA ; 317(18): 1844-1853, 2017 05 09.
Article de Anglais | MEDLINE | ID: mdl-28492898

RÉSUMÉ

Importance: There are no specifically approved targeted therapies for the most common genomically defined subset of non-small cell lung cancer (NSCLC), KRAS-mutant lung cancer. Objective: To compare efficacy of the mitogen-activated protein kinase kinase (MEK) inhibitor selumetinib + docetaxel with docetaxel alone as a second-line therapy for advanced KRAS-mutant NSCLC. Design, Setting, and Participants: Multinational, randomized clinical trial conducted at 202 sites across 25 countries from October 2013 through January 2016. Of 3323 patients with advanced NSCLC and disease progression following first-line anticancer therapy tested for a KRAS mutation, 866 were enrolled and 510 randomized. Primary reason for exclusion was ineligibility. The data cutoff date for analysis was June 7, 2016. Interventions: Patients were randomized 1:1; 254 to receive selumetinib + docetaxel and 256 to receive placebo + docetaxel. Main Outcomes and Measures: Primary end point was investigator assessed progression-free survival. Secondary end points included overall survival, objective response rate, duration of response, effects on disease-related symptoms, safety, and tolerability. Results: Of 510 randomized patients (mean age, 61.4 years [SD, 8.3]; women, 207 [41%]), 505 patients (99%) received treatment and completed the study (251 received selumetinib + docetaxel; 254 received placebo + docetaxel). At the time of data cutoff, 447 patients (88%) had experienced a progression event and 346 deaths (68%) had occurred. Median progression-free survival was 3.9 months (interquartile range [IQR], 1.5-5.9) with selumetinib + docetaxel and 2.8 months (IQR, 1.4-5.5) with placebo + docetaxel (difference, 1.1 months; hazard ratio [HR], 0.93 [95% CI, 0.77-1.12]; P = .44). Median overall survival was 8.7 months (IQR, 3.6-16.8) with selumetinib + docetaxel and 7.9 months (IQR, 3.8-20.1) with placebo + docetaxel (difference, 0.9 months; HR, 1.05 [95% CI, 0.85-1.30]; P = .64). Objective response rate was 20.1% with selumetinib + docetaxel and 13.7% with placebo + docetaxel (difference, 6.4%; odds ratio, 1.61 [95% CI, 1.00-2.62]; P = .05). Median duration of response was 2.9 months (IQR, 1.7-4.8; 95% CI, 2.7-4.1) with selumetinib + docetaxel and 4.5 months (IQR, 2.3-7.3; 95% CI, 2.8-5.6) with placebo + docetaxel. Adverse events of grade 3 or higher were more frequent with selumetinib + docetaxel (169 adverse events [67%] for selumetinib + docetaxel vs 115 adverse events [45%] for placebo + docetaxel; difference, 22%). Conclusions and Relevance: Among patients with previously treated advanced KRAS-mutant non-small cell lung cancer, addition of selumetinib to docetaxel did not improve progression-free survival compared with docetaxel alone. Trial Registration: clinicaltrials.gov: NCT01933932.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Benzimidazoles/administration et posologie , Protéines proto-oncogènes p21(ras)/génétique , Taxoïdes/administration et posologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Benzimidazoles/effets indésirables , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Évolution de la maladie , Survie sans rechute , Docetaxel , Femelle , Humains , Mâle , Adulte d'âge moyen , Mutation , Taxoïdes/effets indésirables , Résultat thérapeutique
12.
Oncotarget ; 8(64): 107492-107499, 2017 Dec 08.
Article de Anglais | MEDLINE | ID: mdl-29296181

RÉSUMÉ

MEK inhibitor (selumetinib) is a potent, orally active inhibitor of MAPK/ERK pathway. It is important to develop an accurate and robust method indicative of RAS pathway activity to stratify potential patients who can benefit from selumetinib treatment in gastric cancer (GC). First, we surveyed the sensitivity to selumetinib in a panel of 22 GC cell lines and correlated with MEK signature to selumetinib sensitivity. Next, we analyzed MEK signature via nanostring assay in two Asian cohorts using clinical samples (n = 218) and then performed a correlative analysis with MEK signature status and KRAS genotype in GC. MEK signature was predictive of response of selumetinib in GC cell lines regardless of KRAS mutation status. The proportion of high MEK signature by nanostring assay was 6.9% and the proportion of high MEK signature was significantly higher in KRAS altered group in a Korean cohort. None of PIK3CA altered cases belonged to high MEK signature group. MEK high signature was more prevalent in intestinal type by Lauren classification. The correlation between MEK signature, KRAS alteration and treatment response to selumetinib should be validated in prospective clinical trials.

13.
Mol Cancer Ther ; 15(11): 2802-2813, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27550940

RÉSUMÉ

The challenge of developing effective pharmacodynamic biomarkers for preclinical and clinical testing of FGFR signaling inhibition is significant. Assays that rely on the measurement of phospho-protein epitopes can be limited by the availability of effective antibody detection reagents. Transcript profiling enables accurate quantification of many biomarkers and provides a broader representation of pathway modulation. To identify dynamic transcript biomarkers of FGFR signaling inhibition by AZD4547, a potent inhibitor of FGF receptors 1, 2, and 3, a gene expression profiling study was performed in FGFR2-amplified, drug-sensitive tumor cell lines. Consistent with known signaling pathways activated by FGFR, we identified transcript biomarkers downstream of the RAS-MAPK and PI3K/AKT pathways. Using different tumor cell lines in vitro and xenografts in vivo, we confirmed that some of these transcript biomarkers (DUSP6, ETV5, YPEL2) were modulated downstream of oncogenic FGFR1, 2, 3, whereas others showed selective modulation only by FGFR2 signaling (EGR1). These transcripts showed consistent time-dependent modulation, corresponding to the plasma exposure of AZD4547 and inhibition of phosphorylation of the downstream signaling molecules FRS2 or ERK. Combination of FGFR and AKT inhibition in an FGFR2-mutated endometrial cancer xenograft model enhanced modulation of transcript biomarkers from the PI3K/AKT pathway and tumor growth inhibition. These biomarkers were detected on the clinically validated nanoString platform. Taken together, these data identified novel dynamic transcript biomarkers of FGFR inhibition that were validated in a number of in vivo models, and which are more robustly modulated by FGFR inhibition than some conventional downstream signaling protein biomarkers. Mol Cancer Ther; 15(11); 2802-13. ©2016 AACR.


Sujet(s)
Antinéoplasiques/pharmacologie , Benzamides/pharmacologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Pipérazines/pharmacologie , Pyrazoles/pharmacologie , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Transcriptome , Animaux , Marqueurs biologiques , Lignée cellulaire tumorale , Analyse de regroupements , Modèles animaux de maladie humaine , Femelle , Amplification de gène , Analyse de profil d'expression de gènes , Hétérogreffes , Humains , Souris , Récepteur facteur croissance fibroblaste/génétique , Récepteur facteur croissance fibroblaste/métabolisme , Reproductibilité des résultats , Transduction du signal/effets des médicaments et des substances chimiques
14.
Cancer Discov ; 6(8): 838-851, 2016 08.
Article de Anglais | MEDLINE | ID: mdl-27179038

RÉSUMÉ

UNLABELLED: FGFR1 and FGFR2 are amplified in many tumor types, yet what determines response to FGFR inhibition in amplified cancers is unknown. In a translational clinical trial, we show that gastric cancers with high-level clonal FGFR2 amplification have a high response rate to the selective FGFR inhibitor AZD4547, whereas cancers with subclonal or low-level amplification did not respond. Using cell lines and patient-derived xenograft models, we show that high-level FGFR2 amplification initiates a distinct oncogene addiction phenotype, characterized by FGFR2-mediated transactivation of alternative receptor kinases, bringing PI3K/mTOR signaling under FGFR control. Signaling in low-level FGFR1-amplified cancers is more restricted to MAPK signaling, limiting sensitivity to FGFR inhibition. Finally, we show that circulating tumor DNA screening can identify high-level clonally amplified cancers. Our data provide a mechanistic understanding of the distinct pattern of oncogene addiction seen in highly amplified cancers and demonstrate the importance of clonality in predicting response to targeted therapy. SIGNIFICANCE: Robust single-agent response to FGFR inhibition is seen only in high-level FGFR-amplified cancers, with copy-number level dictating response to FGFR inhibition in vitro, in vivo, and in the clinic. High-level amplification of FGFR2 is relatively rare in gastric and breast cancers, and we show that screening for amplification in circulating tumor DNA may present a viable strategy to screen patients. Cancer Discov; 6(8); 838-51. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 803.


Sujet(s)
Antinéoplasiques/pharmacologie , Benzamides/pharmacologie , Évolution clonale/génétique , Amplification de gène , Pipérazines/pharmacologie , Pyrazoles/pharmacologie , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Récepteur facteur croissance fibroblaste/génétique , Animaux , Tumeurs du sein/diagnostic , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Lignée cellulaire tumorale , Femelle , Analyse de profil d'expression de gènes , Humains , Mâle , Souris , Thérapie moléculaire ciblée , Phosphatidylinositol 3-kinases/métabolisme , Phosphorylation , Tomographie par émission de positons , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs de l'estomac/diagnostic , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/génétique , Tachykinines/métabolisme , Tomodensitométrie , Tests d'activité antitumorale sur modèle de xénogreffe
15.
PLoS One ; 11(2): e0149628, 2016.
Article de Anglais | MEDLINE | ID: mdl-26905262

RÉSUMÉ

FGFR1 amplification occurs in ~20% of sqNSCLC and trials with FGFR inhibitors have selected FGFR1 amplified patients by FISH. Lung cancer cell lines were profiled for sensitivity to AZD4547, a potent, selective inhibitor of FGFRs 1-3. Sensitivity to FGFR inhibition was associated with but not wholly predicted by increased FGFR1 gene copy number. Additional biomarker assays evaluating expression of FGFRs and correlation between amplification and expression in clinical tissues are therefore warranted. We validated nanoString for mRNA expression analysis of 194 genes, including FGFRs, from clinical tumour tissue. In a panel of sqNSCLC tumours 14.4% (13/90) were FGFR1 amplified by FISH. Although mean FGFR1 expression was significantly higher in amplified samples, there was significant overlap in the range of expression levels between the amplified and non-amplified cohorts with several non-amplified samples expressing FGFR1 to levels equivalent to amplified samples. Statistical analysis revealed increased expression of FGFR1 neighboring genes on the 8p12 amplicon (BAG4, LSM1 and WHSC1L1) in FGFR1 amplified tumours, suggesting a broad rather than focal amplicon and raises the potential for codependencies. High resolution aCGH analysis of pre-clinical and clinical samples supported the presence of a broad and heterogeneous amplicon around the FGFR1 locus. In conclusion, the range of FGFR1 expression levels in both FGFR1 amplified and non-amplified NSCLC tissues, together with the breadth and intra-patient heterogeneity of the 8p amplicon highlights the need for gene expression analysis of clinical samples to inform the understanding of determinants of response to FGFR inhibitors. In this respect the nanoString platform provides an attractive option for RNA analysis of FFPE clinical samples.


Sujet(s)
Benzamides/pharmacologie , Carcinome pulmonaire non à petites cellules/génétique , Tumeurs du poumon/génétique , Thérapie moléculaire ciblée/méthodes , Pipérazines/pharmacologie , Pyrazoles/pharmacologie , Récepteur FGFR1/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Lignée cellulaire tumorale/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Chromosomes humains de la paire 8 , Hybridation génomique comparative , Amplification de gène , Régulation de l'expression des gènes tumoraux , Humains , Hybridation fluorescente in situ , Tumeurs du poumon/traitement médicamenteux , Reproductibilité des résultats
16.
J Hematol Oncol ; 8: 119, 2015 Oct 24.
Article de Anglais | MEDLINE | ID: mdl-26497743

RÉSUMÉ

BACKGROUND: Urothelial cancers (UC) are the fourth most common tumours worldwide after prostate (or breast), lung and colorectal cancer. Despite recent improvements in their management, UC remain an aggressive disease associated with a poor outcome. Following disease progression on first-line platinum-based chemotherapy, very few effective treatment options are available and none of them have shown significant improvement in overall survival. Alterations of the fibroblast growth factor receptor (FGFR) pathway including amplification, mutations and overexpression are common in UC. Pre-clinical data suggest that the presence of such dysregulations may confer sensitivity to FGFR inhibitors. MATERIALS AND METHODS: We present here the case of a patient with a metastatic UC of the renal pelvis with lymph node metastases treated with the selective FGFR inhibitor AZD4547. RESULTS: To date, the patient has been on a study drug for 32 months with acceptable tolerance and maintained radiological partial response as per RECIST 1.1 criteria. Exploratory biomarker analysis showed FGFR3, FGFR1, FGF-ligand and fibroblast growth factor receptor substrate 2 (FRS2) expression in the patient's tumour, together with the presence of a germ-line mutation in the FGFR3 extracellular binding domain. This is not a known hotspot mutation, and the functional significance remains unclear. CONCLUSIONS: The FGFR inhibitor AZD4547 exhibits antitumour activity in a metastatic urothelial cancer displaying FGFR1, FGFR3, FGF-ligand and FRS2 expression. This lends support to the further exploration of FGFR inhibitors in urothelial cancer. Further studies are required to determinate the most effective way to select those patients most likely to respond.


Sujet(s)
Récepteurs ErbB/génétique , Transduction du signal/génétique , Tumeurs de l'uretère/génétique , Tumeurs de la vessie urinaire/génétique , Antinéoplasiques/usage thérapeutique , Benzamides/usage thérapeutique , Récepteurs ErbB/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Immunohistochimie , Hybridation fluorescente in situ , Pelvis rénal/métabolisme , Pelvis rénal/anatomopathologie , Mâle , Adulte d'âge moyen , Métastase tumorale , Pipérazines/usage thérapeutique , Pyrazoles/usage thérapeutique , Récepteur FGFR1/génétique , Récepteur FGFR1/métabolisme , Récepteur de type 3 des facteurs de croissance fibroblastique/génétique , Récepteur de type 3 des facteurs de croissance fibroblastique/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs de l'uretère/traitement médicamenteux , Tumeurs de l'uretère/métabolisme , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/métabolisme
17.
PLoS One ; 8(6): e66963, 2013.
Article de Anglais | MEDLINE | ID: mdl-23826179

RÉSUMÉ

Phase III trials of the anti-insulin-like growth factor-1 receptor (IGF1R) antibody figitumumab in non-small cell lung cancer (NSCLC) patients have been discontinued owing to lack of survival benefit. We investigated whether inhibition of the highly homologous insulin receptor (IR) in addition to the IGF1R would be more effective than inhibition of the IGF1R alone at preventing the proliferation of NSCLC cells. Signalling through IGF1R and IR in the NSCLC cell lines A549 and Hcc193 was stimulated by a combination of IGF1, IGF2 and insulin. It was inhibited by antibodies that block ligand binding, αIR3 (IGF1R) and IR47-9 (IR), and by the ATP-competitive small molecule tyrosine kinase inhibitors AZ12253801 and NVPAWD742 which inhibit both IGF1R and IR tyrosine kinases. The effect of inhibitors was determined by an anchorage-independent proliferation assay and by analysis of Akt phosphorylation. In Hcc193 cells the reduction in cell proliferation and Akt phosphorylation due to anti-IGF1R antibody was enhanced by antibody-mediated inhibition of the IR whereas in A549 cells, with a relatively low IR:IGF1R expression ratio, it was not. In each cell line proliferation and Akt phosphorylation were more effectively inhibited by AZ12253801 and NVPAWD742 than by combined αIR3 and IR47-9. When the IGF1R alone is inhibited, unencumbered signalling through the IR can contribute to continued NSCLC cell proliferation. We conclude that small molecule inhibitors targeting both the IR and IGF1R more effectively reduce NSCLC cell proliferation in a manner independent of the IR:IGF1R expression ratio, providing a therapeutic rationale for the treatment of this disease.


Sujet(s)
Carcinome pulmonaire non à petites cellules/métabolisme , Tumeurs du poumon/métabolisme , Thérapie moléculaire ciblée , Récepteur IGF de type 1/antagonistes et inhibiteurs , Récepteur à l'insuline/antagonistes et inhibiteurs , Anticorps monoclonaux/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Humains , Isoxazoles/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Pyrimidines/pharmacologie , Récepteur IGF de type 1/métabolisme , Récepteur à l'insuline/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
18.
Clin Cancer Res ; 19(9): 2572-83, 2013 May 01.
Article de Anglais | MEDLINE | ID: mdl-23493349

RÉSUMÉ

PURPOSE: FGFR gene aberrations are associated with tumor growth and survival. We explored the role of FGFR2 amplification in gastric cancer and the therapeutic potential of AZD4547, a potent and selective ATP-competitive receptor tyrosine kinase inhibitor of fibroblast growth factor receptor (FGFR)1-3, in patients with FGFR2-amplified gastric cancer. EXPERIMENTAL DESIGN: Array-comparative genomic hybridization and FISH were used to identify FGFR2 amplification in gastric cancer patient tumor samples. The effects of FGFR2 modulation were investigated in gastric cancer cells with FGFR2 amplification and in patient-derived gastric cancer xenograft (PDGCX) models using two approaches: inhibition with AZD4547 and short hairpin RNA (shRNA) knockdown of FGFR2. RESULTS: Amplification of the FGFR2 gene was identified in a subset of Chinese and Caucasian patients with gastric cancer. Gastric cancer cell lines SNU-16 and KATOIII, carrying the amplified FGFR2 gene, were extremely sensitive to AZD4547 in vitro with GI50 values of 3 and 5 nmol/L, respectively. AZD4547 effectively inhibited phosphorylation of FGFR2 and its downstream signaling molecules and induced apoptosis in SNU-16 cells. Furthermore, inhibition of FGFR2 signaling by AZD4547 resulted in significant dose-dependent tumor growth inhibition in FGFR2-amplified xenograft (SNU-16) and PDGCX models (SGC083) but not in nonamplified models. shRNA knockdown of FGFR2 similarly inhibited tumor growth in vitro and in vivo. Finally, compared with monotherapy, we showed enhancement of in vivo antitumor efficacy using AZD4547 in combination with chemotherapeutic agents. CONCLUSION: FGFR2 pathway activation is required for driving growth and survival of gastric cancer carrying FGFR2 gene amplification both in vitro and in vivo. Our data support therapeutic intervention with FGFR inhibitors, such as AZD4547, in patients with gastric cancer carrying FGFR2 gene amplification.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Amplification de gène , Récepteur FGFR2/génétique , Tumeurs de l'estomac/génétique , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Apoptose , Benzamides/administration et posologie , Camptothécine/administration et posologie , Camptothécine/analogues et dérivés , Études cas-témoins , Lignée cellulaire tumorale , Cisplatine/administration et posologie , Docetaxel , Synergie des médicaments , Femelle , Fluorouracil/administration et posologie , Techniques de knock-down de gènes , Humains , Concentration inhibitrice 50 , Irinotécan , Mâle , Souris , Souris de lignée BALB C , Souris nude , Adulte d'âge moyen , Pipérazines/administration et posologie , Pyrazoles/administration et posologie , Petit ARN interférent/génétique , Récepteur FGFR2/antagonistes et inhibiteurs , Récepteur FGFR2/métabolisme , Transduction du signal , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/métabolisme , Taxoïdes/administration et posologie , Résultat thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe , Jeune adulte
19.
J Med Chem ; 55(22): 10136-47, 2012 Nov 26.
Article de Anglais | MEDLINE | ID: mdl-23088558

RÉSUMÉ

Inhibition of 11ß-HSD1 is viewed as a potential target for the treatment of obesity and other elements of the metabolic syndrome. We report here the optimization of a carboxylic acid class of inhibitors from AZD4017 (1) to the development candidate AZD8329 (27). A structural change from pyridine to pyrazole together with structural optimization led to an improved technical profile in terms of both solubility and pharmacokinetics. The extent of acyl glucuronidation was reduced through structural optimization of both the carboxylic acid and amide substituents, coupled with a reduction in lipophilicity leading to an overall increase in metabolic stability.


Sujet(s)
11-beta-Hydroxysteroid dehydrogenase type 1/antagonistes et inhibiteurs , Benzoates/pharmacologie , Antienzymes/pharmacologie , Glucuronides/métabolisme , Pyrazoles/composition chimique , Pyrazoles/pharmacologie , Pyridines/composition chimique , 11-beta-Hydroxysteroid dehydrogenase type 1/métabolisme , Tissu adipeux/effets des médicaments et des substances chimiques , Tissu adipeux/enzymologie , Animaux , Benzoates/synthèse chimique , Benzoates/pharmacocinétique , Chiens , Antienzymes/synthèse chimique , Antienzymes/pharmacocinétique , Glucuronides/composition chimique , Cochons d'Inde , Humains , Foie/effets des médicaments et des substances chimiques , Foie/enzymologie , Macaca fascicularis , Souris , Modèles moléculaires , Structure moléculaire , Conformation des protéines , Pyrazoles/synthèse chimique , Pyrazoles/pharmacocinétique , Rats , Rat Wistar , Relation structure-activité , Spécificité du substrat
20.
Clin Cancer Res ; 18(24): 6658-67, 2012 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-23082000

RÉSUMÉ

PURPOSE: To investigate the incidence of FGFR1 amplification in Chinese non-small cell lung cancer (NSCLC) and to preclinically test the hypothesis that the novel, potent, and selective fibroblast growth factor receptor (FGFR) small-molecule inhibitor AZD4547 will deliver potent antitumor activity in NSCLC FGFR1-amplified patient-derived tumor xenograft (PDTX) models. EXPERIMENTAL DESIGN: A range of assays was used to assess the translational relevance of FGFR1 amplification and AZD4547 treatment including in vitro lung cell line panel screening and pharmacodynamic (PD) analysis, FGFR1 FISH tissue microarray (TMA) analysis of Chinese NSCLC (n = 127), and, importantly, antitumor efficacy testing and PD analysis of lung PDTX models using AZD4547. RESULTS: The incidence of FGFR1 amplification within Chinese patient NSCLC tumors was 12.5% of squamous origin (6 of 48) and 7% of adenocarcinoma (5 of 76). AZD4547 displayed a highly selective profile across a lung cell line panel, potently inhibiting cell growth only in those lines harboring amplified FGFR1 (GI(50) = 0.003-0.111 µmol/L). AZD4547 induced potent tumor stasis or regressive effects in four of five FGFR1-amplified squamous NSCLC PDTX models. Pharmacodynamic modulation was observed in vivo, and antitumor efficacy correlated well with FGFR1 FISH score and protein expression level. CONCLUSIONS: This study provides novel epidemiologic data through identification of FGFR1 gene amplification in Chinese NSCLC specimens (particularly squamous) and, importantly, extends the clinical significance of this finding by using multiple FGFR1-amplified squamous lung cancer PDTX models to show tumor stasis or regression effects using a specific FGFR inhibitor (AZD4547). Thus, the translational science presented here provides a strong rationale for investigation of AZD4547 as a therapeutic option for patients with squamous NSCLC tumors harboring amplification of FGFR1.


Sujet(s)
Antinéoplasiques/pharmacologie , Benzamides/pharmacologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/traitement médicamenteux , Pipérazines/pharmacologie , Pyrazoles/pharmacologie , Récepteur FGFR1/génétique , Animaux , Antinéoplasiques/usage thérapeutique , Benzamides/usage thérapeutique , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Amplification de gène , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Mâle , Souris , Souris nude , Pipérazines/usage thérapeutique , Pyrazoles/usage thérapeutique , Récepteur FGFR1/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...