Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Nat Genet ; 52(12): 1373-1383, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33230298

RESUMEN

We performed harmonized molecular and clinical analysis on 1,048 melanomas and discovered markedly different global genomic properties among subtypes (BRAF, (N)RAS, NF1, triple wild-type (TWT)), subtype-specific preferences for secondary driver genes and active mutational processes previously unreported in melanoma. Secondary driver genes significantly enriched in specific subtypes reflected preferential dysregulation of additional pathways, such as induction of transforming growth factor-ß signaling in BRAF melanomas and inactivation of the SWI/SNF complex in (N)RAS melanomas, and select co-mutation patterns coordinated selective response to immune checkpoint blockade. We also defined the mutational landscape of TWT melanomas and revealed enrichment of DNA-repair-defect signatures in this subtype, which were associated with transcriptional downregulation of key DNA-repair genes, and may revive previously discarded or currently unconsidered therapeutic modalities for genomically stratified melanoma patient subsets. Broadly, harmonized meta-analysis of melanoma whole exomes revealed distinct molecular drivers that may point to multiple opportunities for biological and therapeutic investigation.


Asunto(s)
Trastornos por Deficiencias en la Reparación del ADN/genética , GTP Fosfohidrolasas/genética , Melanoma/genética , Proteínas de la Membrana/genética , Neurofibromina 1/genética , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/genética , Reparación del ADN/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Melanoma/patología , Transducción de Señal/genética , Neoplasias Cutáneas/patología , Secuenciación del Exoma
3.
Cancer Immunol Res ; 8(8): 1075-1084, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32321774

RESUMEN

Prior data have variably implicated the inactivation of the mammalian SWItch/Sucrose Non-Fermentable (mSWI/SNF) complex with increased tumor sensitivity to immune checkpoint inhibitors (ICI). Herein, we examined the association between mSWI/SNF variants and clinical outcomes to ICIs. We correlated somatic loss-of-function (LOF) variants in a predefined set of mSWI/SNF genes (ARID1A, ARID1B, SMARCA4, SMARCB1, PBRM1, and ARID2) with clinical outcomes in patients with cancer treated with systemic ICIs. We identified 676 patients from Dana-Farber Cancer Institute (DFCI, Boston, MA) and 848 patients from a publicly available database from Memorial Sloan Kettering Cancer Center (MSKCC, New York, NY) who met the inclusion criteria. Multivariable analyses were conducted and adjusted for available baseline factors and tumor mutational burden. Median follow-up was 19.6 (17.6-22.0) months and 28.0 (25.0-29.0) months for the DFCI and MSKCC cohorts, respectively. Seven solid tumor subtypes were examined. In the DFCI cohort, LOF variants of mSWI/SNF did not predict improved overall survival (OS), time-to-treatment failure (TTF), or disease control rate. Only patients with renal cell carcinoma with mSWI/SNF LOF showed significantly improved OS and TTF with adjusted HRs (95% confidence interval) of 0.33 (0.16-0.7) and 0.49 (0.27-0.88), respectively, and this was mostly driven by PRBM1 In the MSKCC cohort, where only OS was captured, LOF mSWI/SNF did not correlate with improved outcomes across any tumor subtype. We did not find a consistent association between mSWI/SNF LOF variants and improved clinical outcomes to ICIs, suggesting that mSWI/SNF variants should not be considered as biomarkers of response to ICIs.


Asunto(s)
Biomarcadores de Tumor/genética , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Mutación , Neoplasias/genética , Neoplasias/terapia , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/inmunología , Estudios de Cohortes , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Humanos , Persona de Mediana Edad , Neoplasias/inmunología , Proteínas Nucleares/genética , Proteína SMARCB1/genética , Tasa de Supervivencia , Factores de Transcripción/genética , Adulto Joven
4.
Clin Cancer Res ; 25(18): 5561-5571, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31253631

RESUMEN

PURPOSE: Molecular properties associated with complete response or acquired resistance to concurrent chemotherapy and radiotherapy (CRT) are incompletely characterized.Experimental Design: We performed integrated whole-exome/transcriptome sequencing and immune infiltrate analysis on rectal adenocarcinoma tumors prior to neoadjuvant CRT (pre-CRT) and at time of resection (post-CRT) in 17 patients [8 complete/partial responders, 9 nonresponders (NR)]. RESULTS: CRT was not associated with increased tumor mutational burden or neoantigen load and did not alter the distribution of established somatic tumor mutations in rectal cancer. Concurrent KRAS/TP53 mutations (KP) associated with NR tumors and were enriched for an epithelial-mesenchymal transition transcriptional program. Furthermore, NR was associated with reduced CD4/CD8 T-cell infiltrates and a post-CRT M2 macrophage phenotype. Absence of any local tumor recurrences, KP/NR status predicted worse progression-free survival, suggesting that local immune escape during or after CRT with specific genomic features contributes to distant progression. CONCLUSIONS: Overall, while CRT did not impact genomic profiles, CRT impacted the tumor immune microenvironment, particularly in resistant cases.


Asunto(s)
Biomarcadores de Tumor , Resistencia a Antineoplásicos , Tolerancia a Radiación , Neoplasias del Recto/etiología , Antígenos de Neoplasias/inmunología , Quimioradioterapia Adyuvante , Resistencia a Antineoplásicos/genética , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Tolerancia a Radiación/genética , Neoplasias del Recto/metabolismo , Neoplasias del Recto/terapia , Transcriptoma , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Secuenciación del Exoma
5.
Artículo en Inglés | MEDLINE | ID: mdl-31832578

RESUMEN

PURPOSE: Heterogeneity in tumor mutational burden (TMB) quantification across sequencing platforms limits the application and further study of this potential biomarker of response to immune checkpoint inhibitors (ICI). We hypothesized that harmonization of TMB across platforms would enable integration of distinct clinical datasets to better characterize the association between TMB and ICI response. METHODS: Cohorts of NSCLC patients sequenced by one of three targeted panels or by whole exome sequencing (WES) were compared (total n=7297). TMB was calculated uniformly and compared across cohorts. TMB distributions were harmonized by applying a normal transformation followed by standardization to z-scores. In sub-cohorts of patients treated with ICIs (DFCI n=272; MSKCC n=227), the association between TMB and outcome was assessed. Durable clinical benefit (DCB) was defined as responsive/stable disease lasting ≥6 months. RESULTS: TMB values were higher in the panel cohorts than the WES cohort. Average mutation rates per gene were highly concordant across cohorts (Pearson coefficient 0.842-0.866). Subsetting the WES cohort by gene panels only partially reproduced the observed differences in TMB. Standardization of TMB into z-scores harmonized TMB distributions and enabled integration of the ICI-treated sub-cohorts. Simulations indicated that cohorts >900 are necessary for this approach. TMB did not associate with response in never smokers or patients harboring targetable driver alterations, although these analyses were under-powered. Increasing TMB thresholds increased DCB rate, but DCB rates within deciles varied. Receiver operator curves yielded an area under the curve of 0.614 with no natural inflection point. CONCLUSION: Z-score conversion harmonizes TMB values and enables integration of datasets derived from different sequencing panels. Clinical and biologic features may provide context to the clinical application of TMB, and warrant further study.

6.
Nat Med ; 25(12): 1916-1927, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31792460

RESUMEN

Immune-checkpoint blockade (ICB) has demonstrated efficacy in many tumor types, but predictors of responsiveness to anti-PD1 ICB are incompletely characterized. In this study, we analyzed a clinically annotated cohort of patients with melanoma (n = 144) treated with anti-PD1 ICB, with whole-exome and whole-transcriptome sequencing of pre-treatment tumors. We found that tumor mutational burden as a predictor of response was confounded by melanoma subtype, whereas multiple novel genomic and transcriptomic features predicted selective response, including features associated with MHC-I and MHC-II antigen presentation. Furthermore, previous anti-CTLA4 ICB exposure was associated with different predictors of response compared to tumors that were naive to ICB, suggesting selective immune effects of previous exposure to anti-CTLA4 ICB. Finally, we developed parsimonious models integrating clinical, genomic and transcriptomic features to predict intrinsic resistance to anti-PD1 ICB in individual tumors, with validation in smaller independent cohorts limited by the availability of comprehensive data. Broadly, we present a framework to discover predictive features and build models of ICB therapeutic response.


Asunto(s)
Antígeno CTLA-4/inmunología , Melanoma/tratamiento farmacológico , Melanoma/genética , Receptor de Muerte Celular Programada 1/inmunología , Anticuerpos Monoclonales Humanizados/administración & dosificación , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Masculino , Melanoma/inmunología , Melanoma/patología , Mutación/genética , Metástasis de la Neoplasia , Nivolumab/administración & dosificación , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Transcriptoma/genética , Transcriptoma/inmunología , Secuenciación del Exoma
7.
Nat Biotechnol ; 36(11): 1056-1058, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30114007

RESUMEN

We present an in silico approach to identifying neoepitopes derived from intron retention events in tumor transcriptomes. Using mass spectrometry immunopeptidome analysis, we show that retained intron neoepitopes are processed and presented on MHC I on the surface of cancer cell lines. RNA-derived neoepitopes should be considered for prospective personalized cancer vaccine development.


Asunto(s)
Simulación por Computador , Epítopos/genética , Intrones/genética , Modelos Genéticos , Neoplasias/genética , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Mapeo Epitopo , Epítopos/metabolismo , Humanos , Neoplasias/inmunología , Neoplasias/terapia , ARN/genética
8.
Science ; 359(6377): 801-806, 2018 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-29301960

RESUMEN

Immune checkpoint inhibitors targeting the programmed cell death 1 receptor (PD-1) improve survival in a subset of patients with clear cell renal cell carcinoma (ccRCC). To identify genomic alterations in ccRCC that correlate with response to anti-PD-1 monotherapy, we performed whole-exome sequencing of metastatic ccRCC from 35 patients. We found that clinical benefit was associated with loss-of-function mutations in the PBRM1 gene (P = 0.012), which encodes a subunit of the PBAF switch-sucrose nonfermentable (SWI/SNF) chromatin remodeling complex. We confirmed this finding in an independent validation cohort of 63 ccRCC patients treated with PD-1 or PD-L1 (PD-1 ligand) blockade therapy alone or in combination with anti-CTLA-4 (cytotoxic T lymphocyte-associated protein 4) therapies (P = 0.0071). Gene-expression analysis of PBAF-deficient ccRCC cell lines and PBRM1-deficient tumors revealed altered transcriptional output in JAK-STAT (Janus kinase-signal transducers and activators of transcription), hypoxia, and immune signaling pathways. PBRM1 loss in ccRCC may alter global tumor-cell expression profiles to influence responsiveness to immune checkpoint therapy.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/terapia , Inmunoterapia/métodos , Neoplasias Renales/genética , Neoplasias Renales/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Antígeno CTLA-4/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/genética , Estudios de Cohortes , Exoma/genética , Perfilación de la Expresión Génica , Genómica , Humanos , Mutación , Factores de Transcripción/genética
9.
Nat Genet ; 50(9): 1271-1281, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30150660

RESUMEN

Tumor mutational burden correlates with response to immune checkpoint blockade in multiple solid tumors, although in microsatellite-stable tumors this association is of uncertain clinical utility. Here we uniformly analyzed whole-exome sequencing (WES) of 249 tumors and matched normal tissue from patients with clinically annotated outcomes to immune checkpoint therapy, including radiographic response, across multiple cancer types to examine additional tumor genomic features that contribute to selective response. Our analyses identified genomic correlates of response beyond mutational burden, including somatic events in individual driver genes, certain global mutational signatures, and specific HLA-restricted neoantigens. However, these features were often interrelated, highlighting the complexity of identifying genetic driver events that generate an immunoresponsive tumor environment. This study lays a path forward in analyzing large clinical cohorts in an integrated and multifaceted manner to enhance the ability to discover clinically meaningful predictive features of response to immune checkpoint blockade.


Asunto(s)
Repeticiones de Microsatélite , Neoplasias/genética , Neoplasias/inmunología , Estudios de Cohortes , Exoma , Genómica/métodos , Humanos , Inmunidad/genética , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA