Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
1.
bioRxiv ; 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38798673

RÉSUMÉ

Tumors frequently harbor isogenic yet epigenetically distinct subpopulations of multi-potent cells with high tumor-initiating potential-often called Cancer Stem-Like Cells (CSLCs). These can display preferential resistance to standard-of-care chemotherapy. Single-cell analyses can help elucidate Master Regulator (MR) proteins responsible for governing the transcriptional state of these cells, thus revealing complementary dependencies that may be leveraged via combination therapy. Interrogation of single-cell RNA sequencing profiles from seven metastatic breast cancer patients, using perturbational profiles of clinically relevant drugs, identified drugs predicted to invert the activity of MR proteins governing the transcriptional state of chemoresistant CSLCs, which were then validated by CROP-seq assays. The top drug, the anthelmintic albendazole, depleted this subpopulation in vivo without noticeable cytotoxicity. Moreover, sequential cycles of albendazole and paclitaxel-a commonly used chemotherapeutic -displayed significant synergy in a patient-derived xenograft (PDX) from a TNBC patient, suggesting that network-based approaches can help develop mechanism-based combinatorial therapies targeting complementary subpopulations.

2.
bioRxiv ; 2023 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-37873470

RÉSUMÉ

The Mechanism of Action (MoA) of a drug is generally represented as a small, non-tissue-specific repertoire of high-affinity binding targets. Yet, drug activity and polypharmacology are increasingly associated with a broad range of off-target and tissue-specific effector proteins. To address this challenge, we have implemented an efficient integrative experimental and computational framework leveraging the systematic generation and analysis of drug perturbational profiles representing >700 FDA-approved and experimental oncology drugs, in cell lines selected as high-fidelity models of 23 aggressive tumor subtypes. Protein activity-based analyses revealed highly reproducible, drug-mediated modulation of tissue-specific targets, leading to generation of a proteome-wide polypharmacology map, characterization of MoA-related drug clusters and off-target effects, and identification and experimental validation of novel, tissue-specific inhibitors of undruggable oncoproteins. The proposed framework, which is easily extended to elucidating the MoA of novel small-molecule libraries, could help support more systematic and quantitative approaches to precision oncology.

3.
Cancer Discov ; 13(6): 1386-1407, 2023 06 02.
Article de Anglais | MEDLINE | ID: mdl-37061969

RÉSUMÉ

Predicting in vivo response to antineoplastics remains an elusive challenge. We performed a first-of-kind evaluation of two transcriptome-based precision cancer medicine methodologies to predict tumor sensitivity to a comprehensive repertoire of clinically relevant oncology drugs, whose mechanism of action we experimentally assessed in cognate cell lines. We enrolled patients with histologically distinct, poor-prognosis malignancies who had progressed on multiple therapies, and developed low-passage, patient-derived xenograft models that were used to validate 35 patient-specific drug predictions. Both OncoTarget, which identifies high-affinity inhibitors of individual master regulator (MR) proteins, and OncoTreat, which identifies drugs that invert the transcriptional activity of hyperconnected MR modules, produced highly significant 30-day disease control rates (68% and 91%, respectively). Moreover, of 18 OncoTreat-predicted drugs, 15 induced the predicted MR-module activity inversion in vivo. Predicted drugs significantly outperformed antineoplastic drugs selected as unpredicted controls, suggesting these methods may substantively complement existing precision cancer medicine approaches, as also illustrated by a case study. SIGNIFICANCE: Complementary precision cancer medicine paradigms are needed to broaden the clinical benefit realized through genetic profiling and immunotherapy. In this first-in-class application, we introduce two transcriptome-based tumor-agnostic systems biology tools to predict drug response in vivo. OncoTarget and OncoTreat are scalable for the design of basket and umbrella clinical trials. This article is highlighted in the In This Issue feature, p. 1275.


Sujet(s)
Antinéoplasiques , Tumeurs , Humains , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Transcriptome , Médecine de précision/méthodes , Oncologie médicale/méthodes , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique
4.
Med ; 3(11): 774-791.e7, 2022 11 11.
Article de Anglais | MEDLINE | ID: mdl-36195086

RÉSUMÉ

BACKGROUND: Malignant rhabdoid tumors (MRTs) and Wilms' tumors (WTs) are rare and aggressive renal tumors of infants and young children comprising ∼5% of all pediatric cancers. MRTs are among the most genomically stable cancers, and although WTs are genomically heterogeneous, both generally lack therapeutically targetable genetic mutations. METHODS: Comparative protein activity analysis of MRTs (n = 68) and WTs (n = 132) across TCGA and TARGET cohorts, using metaVIPER, revealed elevated exportin 1 (XPO1) inferred activity. In vitro studies were performed on a panel of MRT and WT cell lines to evaluate effects on proliferation and cell-cycle progression following treatment with the selective XPO1 inhibitor selinexor. In vivo anti-tumor activity was assessed in patient-derived xenograft (PDX) models of MRTs and WTs. FINDINGS: metaVIPER analysis identified markedly aberrant activation of XPO1 in MRTs and WTs compared with other tumor types. All MRT and most WT cell lines demonstrated baseline, aberrant XPO1 activity with in vitro sensitivity to selinexor via cell-cycle arrest and induction of apoptosis. In vivo, XPO1 inhibitors significantly abrogated tumor growth in PDX models, inducing effective disease control with sustained treatment. Corroborating human relevance, we present a case report of a child with multiply relapsed WTs with prolonged disease control on selinexor. CONCLUSIONS: We report on a novel systems-biology-based comparative framework to identify non-genetically encoded vulnerabilities in genomically quiescent pediatric cancers. These results have provided preclinical rationale for investigation of XPO1 inhibitors in an upcoming investigator-initiated clinical trial of selinexor in children with MRTs and WTs and offer opportunities for exploration of inferred XPO1 activity as a potential predictive biomarker for response. FUNDING: This work was funded by CureSearch for Children's Cancer, Alan B. Slifka Foundation, NIH (U01 CA217858, S10 OD012351, and S10 OD021764), Michael's Miracle Cure, Hyundai Hope on Wheels, Cannonball Kids Cancer, Conquer Cancer the ASCO Foundation, Cycle for Survival, Paulie Strong Foundation, and the Grayson Fund.


Sujet(s)
Tumeurs du rein , Enfant , Humains , Enfant d'âge préscolaire , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Tumeurs du rein/traitement médicamenteux ,
6.
Nat Genet ; 50(7): 979-989, 2018 07.
Article de Anglais | MEDLINE | ID: mdl-29915428

RÉSUMÉ

We introduce and validate a new precision oncology framework for the systematic prioritization of drugs targeting mechanistic tumor dependencies in individual patients. Compounds are prioritized on the basis of their ability to invert the concerted activity of master regulator proteins that mechanistically regulate tumor cell state, as assessed from systematic drug perturbation assays. We validated the approach on a cohort of 212 gastroenteropancreatic neuroendocrine tumors (GEP-NETs), a rare malignancy originating in the pancreas and gastrointestinal tract. The analysis identified several master regulator proteins, including key regulators of neuroendocrine lineage progenitor state and immunoevasion, whose role as critical tumor dependencies was experimentally confirmed. Transcriptome analysis of GEP-NET-derived cells, perturbed with a library of 107 compounds, identified the HDAC class I inhibitor entinostat as a potent inhibitor of master regulator activity for 42% of metastatic GEP-NET patients, abrogating tumor growth in vivo. This approach may thus complement current efforts in precision oncology.


Sujet(s)
Antinéoplasiques/pharmacologie , Tumeurs neuroendocrines/traitement médicamenteux , Benzamides/pharmacologie , Lignée cellulaire tumorale , Études de cohortes , Tube digestif/effets des médicaments et des substances chimiques , Tube digestif/métabolisme , Inhibiteurs de désacétylase d'histone/pharmacologie , Histone deacetylases/métabolisme , Humains , Tumeurs de l'intestin/traitement médicamenteux , Tumeurs de l'intestin/génétique , Tumeurs neuroendocrines/génétique , Pancréas/effets des médicaments et des substances chimiques , Pancréas/métabolisme , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/génétique , Médecine de précision/méthodes , Pyridines/pharmacologie , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/génétique
7.
Nat Genet ; 43(9): 830-7, 2011 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-21804550

RÉSUMÉ

Diffuse large B-cell lymphoma (DLBCL) is the most common form of human lymphoma. Although a number of structural alterations have been associated with the pathogenesis of this malignancy, the full spectrum of genetic lesions that are present in the DLBCL genome, and therefore the identity of dysregulated cellular pathways, remains unknown. By combining next-generation sequencing and copy number analysis, we show that the DLBCL coding genome contains, on average, more than 30 clonally represented gene alterations per case. This analysis also revealed mutations in genes not previously implicated in DLBCL pathogenesis, including those regulating chromatin methylation (MLL2; 24% of samples) and immune recognition by T cells. These results provide initial data on the complexity of the DLBCL coding genome and identify novel dysregulated pathways underlying its pathogenesis.


Sujet(s)
Dosage génique , Régulation de l'expression des gènes dans la leucémie , Lymphome B diffus à grandes cellules/génétique , Chromatine/métabolisme , Analyse de mutations d'ADN , Diploïdie , Génome humain , Centre germinatif/immunologie , Humains , Lymphome B diffus à grandes cellules/immunologie , Lymphome B diffus à grandes cellules/anatomopathologie , Méthylation , Récidive tumorale locale/génétique , Récidive tumorale locale/immunologie , Récidive tumorale locale/anatomopathologie , Mutation ponctuelle , Polymorphisme de nucléotide simple , Lymphocytes T/immunologie
8.
J Exp Med ; 208(7): 1389-401, 2011 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-21670202

RÉSUMÉ

The pathogenesis of chronic lymphocytic leukemia (CLL), the most common leukemia in adults, is still largely unknown. The full spectrum of genetic lesions that are present in the CLL genome, and therefore the number and identity of dysregulated cellular pathways, have not been identified. By combining next-generation sequencing and copy number analysis, we show here that the typical CLL coding genome contains <20 clonally represented gene alterations/case, including predominantly nonsilent mutations, and fewer copy number aberrations. These analyses led to the discovery of several genes not previously known to be altered in CLL. Although most of these genes were affected at low frequency in an expanded CLL screening cohort, mutational activation of NOTCH1, observed in 8.3% of CLL at diagnosis, was detected at significantly higher frequency during disease progression toward Richter transformation (31.0%), as well as in chemorefractory CLL (20.8%). Consistent with the association of NOTCH1 mutations with clinically aggressive forms of the disease, NOTCH1 activation at CLL diagnosis emerged as an independent predictor of poor survival. These results provide initial data on the complexity of the CLL coding genome and identify a dysregulated pathway of diagnostic and therapeutic relevance.


Sujet(s)
Leucémie chronique lymphocytaire à cellules B/génétique , Mutation , Récepteur Notch1/génétique , Adulte , Sujet âgé , Évolution de la maladie , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Dosage génique , Régulation de l'expression des gènes tumoraux , Gènes de chaine lourde d'immunoglobuline , Génome humain , Humains , Leucémie chronique lymphocytaire à cellules B/diagnostic , Leucémie chronique lymphocytaire à cellules B/traitement médicamenteux , Mâle , Adulte d'âge moyen , Polymorphisme de nucléotide simple , Pronostic , Échec thérapeutique
9.
Nature ; 471(7337): 189-95, 2011 Mar 10.
Article de Anglais | MEDLINE | ID: mdl-21390126

RÉSUMÉ

B-cell non-Hodgkin's lymphoma comprises biologically and clinically distinct diseases the pathogenesis of which is associated with genetic lesions affecting oncogenes and tumour-suppressor genes. We report here that the two most common types--follicular lymphoma and diffuse large B-cell lymphoma--harbour frequent structural alterations inactivating CREBBP and, more rarely, EP300, two highly related histone and non-histone acetyltransferases (HATs) that act as transcriptional co-activators in multiple signalling pathways. Overall, about 39% of diffuse large B-cell lymphoma and 41% of follicular lymphoma cases display genomic deletions and/or somatic mutations that remove or inactivate the HAT coding domain of these two genes. These lesions usually affect one allele, suggesting that reduction in HAT dosage is important for lymphomagenesis. We demonstrate specific defects in acetylation-mediated inactivation of the BCL6 oncoprotein and activation of the p53 tumour suppressor. These results identify CREBBP/EP300 mutations as a major pathogenetic mechanism shared by common forms of B-cell non-Hodgkin's lymphoma, with direct implications for the use of drugs targeting acetylation/deacetylation mechanisms.


Sujet(s)
Acetyltransferases/génétique , Acetyltransferases/métabolisme , Protéine CBP/génétique , Protéine p300-E1A/génétique , Lymphome B/enzymologie , Lymphome B/génétique , Mutation/génétique , Acétyl coenzyme A/métabolisme , Acétylation , Acetyltransferases/composition chimique , Acetyltransferases/déficit , Animaux , Séquence nucléotidique , Protéine CBP/composition chimique , Protéine CBP/déficit , Protéine CBP/métabolisme , Cellules cultivées , Protéines de liaison à l'ADN/métabolisme , Protéine p300-E1A/composition chimique , Protéine p300-E1A/déficit , Protéine p300-E1A/métabolisme , Régulation de l'expression des gènes tumoraux , Cellules HEK293 , Histone acetyltransferases/composition chimique , Histone acetyltransferases/déficit , Histone acetyltransferases/génétique , Histone acetyltransferases/métabolisme , Humains , Lymphome B/anatomopathologie , Lymphome folliculaire/enzymologie , Lymphome folliculaire/génétique , Lymphome folliculaire/anatomopathologie , Lymphome B diffus à grandes cellules/enzymologie , Lymphome B diffus à grandes cellules/génétique , Lymphome B diffus à grandes cellules/anatomopathologie , Souris , Mutation faux-sens/génétique , Polymorphisme de nucléotide simple/génétique , Liaison aux protéines , Structure tertiaire des protéines/génétique , Protéines proto-oncogènes c-bcl-6 , Récidive , Délétion de séquence/génétique , Protéine p53 suppresseur de tumeur/métabolisme
10.
Nature ; 459(7247): 717-21, 2009 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-19412164

RÉSUMÉ

Diffuse large B-cell lymphoma (DLBCL), the most common form of lymphoma in adulthood, comprises multiple biologically and clinically distinct subtypes including germinal centre B-cell-like (GCB) and activated B-cell-like (ABC) DLBCL. Gene expression profile studies have shown that its most aggressive subtype, ABC-DLBCL, is associated with constitutive activation of the NF-kappaB transcription complex. However, except for a small fraction of cases, it remains unclear whether NF-kappaB activation in these tumours represents an intrinsic program of the tumour cell of origin or a pathogenetic event. Here we show that >50% of ABC-DLBCL and a smaller fraction of GCB-DLBCL carry somatic mutations in multiple genes, including negative (TNFAIP3, also called A20) and positive (CARD11, TRAF2, TRAF5, MAP3K7 (TAK1) and TNFRSF11A (RANK)) regulators of NF-kappaB. Of these, the A20 gene, which encodes a ubiquitin-modifying enzyme involved in termination of NF-kappaB responses, is most commonly affected, with approximately 30% of patients displaying biallelic inactivation by mutations and/or deletions. When reintroduced in cell lines carrying biallelic inactivation of the gene, A20 induced apoptosis and cell growth arrest, indicating a tumour suppressor role. Less frequently, missense mutations of TRAF2 and CARD11 produce molecules with significantly enhanced ability to activate NF-kappaB. Thus, our results demonstrate that NF-kappaB activation in DLBCL is caused by genetic lesions affecting multiple genes, the loss or activation of which may promote lymphomagenesis by leading to abnormally prolonged NF-kappaB responses.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Gènes/génétique , Lymphome B diffus à grandes cellules/génétique , Lymphome B diffus à grandes cellules/physiopathologie , Mutation/génétique , Facteur de transcription NF-kappa B/métabolisme , Apoptose , Lignée cellulaire tumorale , Protéines de liaison à l'ADN , Humains , Protéines et peptides de signalisation intracellulaire/génétique , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Facteur de transcription NF-kappa B/génétique , Protéines nucléaires/génétique , Protéine-3 induite par le facteur de nécrose tumorale alpha
11.
J Exp Med ; 203(2): 311-7, 2006 Feb 20.
Article de Anglais | MEDLINE | ID: mdl-16492805

RÉSUMÉ

PR domain containing 1 with zinc finger domain (PRDM1)/B lymphocyte-induced maturation protein 1 (BLIMP1) is a transcriptional repressor expressed in a subset of germinal center (GC) B cells and in all plasma cells, and required for terminal B cell differentiation. The BLIMP1 locus lies on chromosome 6q21-q22.1, a region frequently deleted in B cell lymphomas, suggesting that it may harbor a tumor suppressor gene. We report here that the BLIMP1 gene is inactivated by structural alterations in 24% (8 out of 34) activated B cell-like diffuse large cell lymphoma (ABC-DLBCL), but not in GC B cell-like (n = 0/37) or unclassified (n = 0/21) DLBCL. BLIMP1 alterations included gene truncations, nonsense mutations, frameshift deletions, and splice site mutations that generate aberrant transcripts encoding truncated BLIMP1 proteins. In all cases studied, both BLIMP1 alleles were inactivated by deletions or mutations. Furthermore, most non-GC type DLBCL cases (n = 20/26, 77%) lack BLIMP1 protein expression, despite the presence of BLIMP1 mRNA. These results indicate that a sizable fraction of ABC-DLBCL carry an inactive BLIMP1 gene, and suggest that the same gene is inactivated by epigenetic mechanisms in an additional large number of cases. These findings point to a role for BLIMP1 as a tumor suppressor gene, whose inactivation may contribute to lymphomagenesis by blocking post-GC differentiation of B cells toward plasma cells.


Sujet(s)
Extinction de l'expression des gènes , Lymphome B/génétique , Lymphome B diffus à grandes cellules/génétique , Protéines de répression/antagonistes et inhibiteurs , Protéines de répression/génétique , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/génétique , Lymphocytes B/anatomopathologie , Différenciation cellulaire/génétique , Lignée cellulaire tumorale , Codon non-sens , Mutation avec décalage du cadre de lecture , Gènes suppresseurs de tumeur , Humains , Lymphome B/classification , Lymphome B/métabolisme , Lymphome B diffus à grandes cellules/classification , Lymphome B diffus à grandes cellules/métabolisme , Plasmocytes/anatomopathologie , Facteur-1 liant le domaine de régulation positive I , Sites d'épissage d'ARN/génétique , Protéines de répression/biosynthèse , Facteurs de transcription/biosynthèse
12.
Am J Hum Genet ; 73(4): 886-97, 2003 Oct.
Article de Anglais | MEDLINE | ID: mdl-13680528

RÉSUMÉ

We previously reported a genomewide scan to identify autism-susceptibility loci in 110 multiplex families, showing suggestive evidence (P <.01) for linkage to autism-spectrum disorders (ASD) on chromosomes 5, 8, 16, 19, and X and showing nominal evidence (P <.05) on several additional chromosomes (2, 3, 4, 10, 11, 12, 15, 18, and 20). In this follow-up analysis we have increased the sample size threefold, while holding the study design constant, so that we now report 345 multiplex families, each with at least two siblings affected with autism or ASD phenotype. Along with 235 new multiplex families, 73 new microsatellite markers were also added in 10 regions, thereby increasing the marker density at these strategic locations from 10 cM to approximately 2 cM and bringing the total number of markers to 408 over the entire genome. Multipoint maximum LOD scores (MLS) obtained from affected-sib-pair analysis of all 345 families yielded suggestive evidence for linkage on chromosomes 17, 5, 11, 4, and 8 (listed in order by MLS) (P <.01). The most significant findings were an MLS of 2.83 (P =.00029) on chromosome 17q, near the serotonin transporter (5-hydroxytryptamine transporter [5-HTT]), and an MLS of 2.54 (P =.00059) on 5p. The present follow-up genome scan, which used a consistent research design across studies and examined the largest ASD sample collection reported to date, gave either equivalent or marginally increased evidence for linkage at several chromosomal regions implicated in our previous scan but eliminated evidence for linkage at other regions.


Sujet(s)
Trouble autistique/génétique , Cartographie chromosomique , Prédisposition génétique à une maladie , Trouble autistique/diagnostic , Famille , Fréquence d'allèle , Dépistage génétique/méthodes , Génome humain , Génotype , Humains , Modèles génétiques , Reproductibilité des résultats
13.
Am J Hum Genet ; 70(1): 51-9, 2002 Jan.
Article de Anglais | MEDLINE | ID: mdl-11715113

RÉSUMÉ

Celiac disease (CD), or gluten-sensitive enteropathy, is a common multifactorial disorder resulting from intolerance to cereal prolamins. The only established genetic susceptibility factor is HLA-DQ, which appears to explain only part of the overall genetic risk. We performed a genomewide scan of CD in 60 Finnish families. In addition to strong evidence for linkage to the HLA region at 6p21.3 (Z(max)>5), suggestive evidence for linkage was found for six other chromosomal regions--1p36, 4p15, 5q31, 7q21, 9p21-23, and 16q12. We further analyzed the three most convincing regions--4p15, 5q31, and 7q21--by evaluation of dense marker arrays across each region and by analysis of an additional 38 families. Although multipoint analysis with dense markers provided supportive evidence (multipoint LOD scores 3.25 at 4p15, 1.49 at 5q31, and 1.04 at 7q21) for the initial findings, the additional 38 families did not strengthen evidence for linkage. The role that HLA-DQ plays was studied in more detail by analysis of DQB1 alleles in all 98 families. All but one patient carried one or two HLA-DQ risk alleles, and 65% of HLA-DQ2 carriers were affected. Our study indicates that the HLA region harbors a predominant CD-susceptibility locus in these Finnish families.


Sujet(s)
Maladie coeliaque/génétique , Liaison génétique/génétique , Prédisposition génétique à une maladie/génétique , Adulte , Allèles , Cartographie chromosomique , Chromosomes humains/génétique , Finlande , Fréquence d'allèle/génétique , Marqueurs génétiques/génétique , Génome humain , Antigènes HLA-DQ/génétique , Chaines bêta des antigènes HLA-DQ , Humains , Lod score
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...