Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
2.
J Minim Invasive Gynecol ; 28(3): 544-555.e7, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33359291

RESUMEN

OBJECTIVE: To compare recurrence rate, progression-free survival (PFS), and overall survival for early-stage cervical cancer after minimally invasive (MIS) vs abdominal radical hysterectomy. DATA SOURCES: MEDLINE, Embase, Cochrane Central Register of Controlled Trials, and Cochrane Library databases. METHODS OF STUDY SELECTION: We identified studies from 1990 to 2020 that included women with stage I or higher cervical cancer treated with primary radical hysterectomy and compared recurrence and/or PFS and overall survival with MIS vs abdominal radical hysterectomy. (The review protocol was registered with the International Prospective Register of Systematic Reviews: CRD4202173600). TABULATION, INTEGRATION, AND RESULTS: We performed random-effects meta-analyses overall and by length of follow-up. Fifty articles on 40 cohort studies and 1 randomized controlled trial that included 22 593 women with cervical cancer met the inclusion criteria. Twenty percent of the studies had <36 months of follow-up, and 24% had more than 60 months of follow-up. The odds of PFS were worse for women undergoing MIS radical hysterectomy (odds ratio 1.54; 95% CI [confidence interval], 1.24-1.94; 14 studies). When limited to studies with longer follow-up, the odds of PFS were progressively worse with MIS radical hysterectomy (HR [hazard ratio] 1.48 for >36 months; 95% CI, 1.21-1.82; 10 studies; HR 1.69 for >48 months; 95% CI, 1.26-2.27; 5 studies; and HR 2.020 for >60 months; 95% CI, 1.36-3.001; 3 studies). For overall survival, the odds were not significantly different for MIS vs abdominal hysterectomy (odds ratio 0.94; 95% CI, 0.66-1.35; 14 studies) (HR 0.99 for >36 months; 95% CI, 0.66-1.48; 9 studies; HR 1.05 for >48 months; 95% CI, 0.57-1.94; 4 studies; and HR 1.35 for >60 months; 95% CI, 0.73-2.51; 3 studies). CONCLUSION: In our meta-analysis of 50 studies, MIS radical hysterectomy was associated with worse PFS than open radical hysterectomy for early-stage cervical cancer. The emergence of this finding with longer follow-up highlights the importance of long-term, high-quality studies to guide cancer and surgical treatments.


Asunto(s)
Histerectomía/métodos , Procedimientos Quirúrgicos Mínimamente Invasivos , Neoplasias del Cuello Uterino/cirugía , Adenocarcinoma/epidemiología , Adenocarcinoma/patología , Adenocarcinoma/cirugía , Adulto , Carcinoma de Células Escamosas/epidemiología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/cirugía , Estudios de Cohortes , Femenino , Humanos , Histerectomía/efectos adversos , Histerectomía/estadística & datos numéricos , Persona de Mediana Edad , Procedimientos Quirúrgicos Mínimamente Invasivos/efectos adversos , Procedimientos Quirúrgicos Mínimamente Invasivos/métodos , Procedimientos Quirúrgicos Mínimamente Invasivos/estadística & datos numéricos , Recurrencia Local de Neoplasia/epidemiología , Estadificación de Neoplasias , Supervivencia sin Progresión , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos , Ensayos Clínicos Controlados Aleatorios como Asunto/estadística & datos numéricos , Análisis de Supervivencia , Neoplasias del Cuello Uterino/epidemiología , Neoplasias del Cuello Uterino/patología
4.
Cancer Discov ; 10(9): 1296-1311, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32371478

RESUMEN

The molecular mechanisms leading to resistance to PD-1 blockade are largely unknown. Here, we characterize tumor biopsies from a patient with melanoma who displayed heterogeneous responses to anti-PD-1 therapy. We observe that a resistant tumor exhibited a loss-of-function mutation in the tumor suppressor gene FBXW7, whereas a sensitive tumor from the same patient did not. Consistent with a functional role in immunotherapy response, inactivation of Fbxw7 in murine tumor cell lines caused resistance to anti-PD-1 in immunocompetent animals. Loss of Fbxw7 was associated with altered immune microenvironment, decreased tumor-intrinsic expression of the double-stranded RNA (dsRNA) sensors MDA5 and RIG1, and diminished induction of type I IFN and MHC-I expression. In contrast, restoration of dsRNA sensing in Fbxw7-deficient cells was sufficient to sensitize them to anti-PD-1. Our results thus establish a new role for the commonly inactivated tumor suppressor FBXW7 in viral sensing and sensitivity to immunotherapy. SIGNIFICANCE: Our findings establish a role of the commonly inactivated tumor suppressor FBXW7 as a genomic driver of response to anti-PD-1 therapy. Fbxw7 loss promotes resistance to anti-PD-1 through the downregulation of viral sensing pathways, suggesting that therapeutic reactivation of these pathways could improve clinical responses to checkpoint inhibitors in genomically defined cancer patient populations.This article is highlighted in the In This Issue feature, p. 1241.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Cutáneas/tratamiento farmacológico , Anciano , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Línea Celular Tumoral/trasplante , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Modelos Animales de Enfermedad , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Regulación Neoplásica de la Expresión Génica/inmunología , Células HeLa , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Helicasa Inducida por Interferón IFIH1/genética , Helicasa Inducida por Interferón IFIH1/metabolismo , Mutación con Pérdida de Función , Masculino , Ratones , Mutagénesis Sitio-Dirigida , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , ARN Bicatenario/inmunología , ARN Bicatenario/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
5.
Gynecol Oncol ; 157(1): 106-114, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31954538

RESUMEN

OBJECTIVE: Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) and SMARCA4-deficient undifferentiated uterine sarcoma (SMARCA4-DUS) are rare and aggressive tumors, primarily affecting pre- and perimenopausal women. Inactivating SMARCA4 mutations are thought to be the driving molecular events in the majority of these tumors. Here, we report the clinical course of a family with germline SMARCA4 mutation and compare large cohorts of these rare tumor types. METHODS: We extracted clinico-pathological medical record data for the family with germline SMARCA4 mutation. Clinico-genomic data from SCCOHT and SMARCA4-DUS cohorts were retrospectively extracted from the archives of a large CLIA-certified reference molecular laboratory. RESULTS: We identified a single family with an inherited germline SMARCA4 mutation, in which two different family members developed either SCCOHT or SMARCA4-DUS, both of whom died within one year of diagnosis, despite aggressive surgical, chemotherapy and immunotherapy treatment. Retrospective comparative analysis of large SCCOHT (n = 48) and SMARCA4-DUS (n = 17) cohorts revealed that SCCOHT patients were younger (median age: 28.5 vs. 49.0) and more likely to have germline SMARCA4 alterations (37.5% vs. 11.8%) than SMARCA4-DUS patients. CONCLUSIONS: Growing understanding of the role SMARCA4 plays in the pathogenesis of these rare cancers may inform recommended genetic testing and counseling in families with these tumor types.


Asunto(s)
Carcinoma de Células Pequeñas/genética , ADN Helicasas/genética , Mutación de Línea Germinal , Hipercalcemia/genética , Proteínas Nucleares/genética , Neoplasias Ováricas/genética , Sarcoma/genética , Factores de Transcripción/genética , Neoplasias Uterinas/genética , Adulto , Carcinoma de Células Pequeñas/patología , Carcinoma de Células Pequeñas/terapia , Diferenciación Celular/fisiología , Estudios de Cohortes , ADN Helicasas/deficiencia , Femenino , Humanos , Hipercalcemia/patología , Hipercalcemia/terapia , Persona de Mediana Edad , Proteínas Nucleares/deficiencia , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Pronóstico , Estudios Retrospectivos , Sarcoma/patología , Sarcoma/terapia , Factores de Transcripción/deficiencia , Neoplasias Uterinas/patología , Neoplasias Uterinas/terapia
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
8.
J Clin Invest ; 128(10): 4441-4453, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30179225

RESUMEN

BACKGROUND: Understanding the integrated immunogenomic landscape of advanced prostate cancer (APC) could impact stratified treatment selection. METHODS: Defective mismatch repair (dMMR) status was determined by either loss of mismatch repair protein expression on IHC or microsatellite instability (MSI) by PCR in 127 APC biopsies from 124 patients (Royal Marsden [RMH] cohort); MSI by targeted panel next-generation sequencing (MSINGS) was then evaluated in the same cohort and in 254 APC samples from the Stand Up To Cancer/Prostate Cancer Foundation (SU2C/PCF). Whole exome sequencing (WES) data from this latter cohort were analyzed for pathogenic MMR gene variants, mutational load, and mutational signatures. Transcriptomic data, available for 168 samples, was also performed. RESULTS: Overall, 8.1% of patients in the RMH cohort had some evidence of dMMR, which associated with decreased overall survival. Higher MSINGS scores associated with dMMR, and these APCs were enriched for higher T cell infiltration and PD-L1 protein expression. Exome MSINGS scores strongly correlated with targeted panel MSINGS scores (r = 0.73, P < 0.0001), and higher MSINGS scores associated with dMMR mutational signatures in APC exomes. dMMR mutational signatures also associated with MMR gene mutations and increased immune cell, immune checkpoint, and T cell-associated transcripts. APC with dMMR mutational signatures overexpressed a variety of immune transcripts, including CD200R1, BTLA, PD-L1, PD-L2, ADORA2A, PIK3CG, and TIGIT. CONCLUSION: These data could impact immune target selection, combination therapeutic strategy selection, and selection of predictive biomarkers for immunotherapy in APC. FUNDING: We acknowledge funding support from Movember, Prostate Cancer UK, The Prostate Cancer Foundation, SU2C, and Cancer Research UK.


Asunto(s)
Antígeno B7-H1 , Reparación de la Incompatibilidad de ADN , Inmunoterapia , Inestabilidad de Microsatélites , Mutación , Proteínas de Neoplasias , Neoplasias de la Próstata , Adulto , Anciano , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia
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
10.
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
11.
Nat Med ; 24(8): 1143-1150, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30038220

RESUMEN

Mesenchymal tumor subpopulations secrete pro-tumorigenic cytokines and promote treatment resistance1-4. This phenomenon has been implicated in chemorefractory small cell lung cancer and resistance to targeted therapies5-8, but remains incompletely defined. Here, we identify a subclass of endogenous retroviruses (ERVs) that engages innate immune signaling in these cells. Stimulated 3 prime antisense retroviral coding sequences (SPARCS) are oriented inversely in 3' untranslated regions of specific genes enriched for regulation by STAT1 and EZH2. Derepression of these loci results in double-stranded RNA generation following IFN-γ exposure due to bi-directional transcription from the STAT1-activated gene promoter and the 5' long terminal repeat of the antisense ERV. Engagement of MAVS and STING activates downstream TBK1, IRF3, and STAT1 signaling, sustaining a positive feedback loop. SPARCS induction in human tumors is tightly associated with major histocompatibility complex class 1 expression, mesenchymal markers, and downregulation of chromatin modifying enzymes, including EZH2. Analysis of cell lines with high inducible SPARCS expression reveals strong association with an AXL/MET-positive mesenchymal cell state. While SPARCS-high tumors are immune infiltrated, they also exhibit multiple features of an immune-suppressed microenviroment. Together, these data unveil a subclass of ERVs whose derepression triggers pathologic innate immune signaling in cancer, with important implications for cancer immunotherapy.


Asunto(s)
Retrovirus Endógenos/metabolismo , Inmunidad Innata/efectos de los fármacos , Interferones/farmacología , Neoplasias/inmunología , Neoplasias/virología , Animales , Línea Celular Tumoral , Retrovirus Endógenos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Desnudos , Neoplasias/genética , ARN sin Sentido/genética
12.
Cell ; 173(3): 624-633.e8, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29656892

RESUMEN

CTLA-4 immune checkpoint blockade is clinically effective in a subset of patients with metastatic melanoma. We identify a subcluster of MAGE-A cancer-germline antigens, located within a narrow 75 kb region of chromosome Xq28, that predicts resistance uniquely to blockade of CTLA-4, but not PD-1. We validate this gene expression signature in an independent anti-CTLA-4-treated cohort and show its specificity to the CTLA-4 pathway with two independent anti-PD-1-treated cohorts. Autophagy, a process critical for optimal anti-cancer immunity, has previously been shown to be suppressed by the MAGE-TRIM28 ubiquitin ligase in vitro. We now show that the expression of the key autophagosome component LC3B and other activators of autophagy are negatively associated with MAGE-A protein levels in human melanomas, including samples from patients with resistance to CTLA-4 blockade. Our findings implicate autophagy suppression in resistance to CTLA-4 blockade in melanoma, suggesting exploitation of autophagy induction for potential therapeutic synergy with CTLA-4 inhibitors.


Asunto(s)
Antígeno CTLA-4/genética , Antígeno CTLA-4/inmunología , Epigénesis Genética , Mutación de Línea Germinal , Neoplasias/genética , Neoplasias/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Autofagia , Línea Celular Tumoral , Metilación de ADN , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunoterapia , Ipilimumab/farmacología , Masculino , Melanoma/genética , Melanoma/inmunología , Antígenos Específicos del Melanoma/genética , Antígenos Específicos del Melanoma/inmunología , Ratones , Ratones Transgénicos , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología
13.
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
14.
Cancer Discov ; 8(2): 196-215, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29101162

RESUMEN

Ex vivo systems that incorporate features of the tumor microenvironment and model the dynamic response to immune checkpoint blockade (ICB) may facilitate efforts in precision immuno-oncology and the development of effective combination therapies. Here, we demonstrate the ability to interrogate ex vivo response to ICB using murine- and patient-derived organotypic tumor spheroids (MDOTS/PDOTS). MDOTS/PDOTS isolated from mouse and human tumors retain autologous lymphoid and myeloid cell populations and respond to ICB in short-term three-dimensional microfluidic culture. Response and resistance to ICB was recapitulated using MDOTS derived from established immunocompetent mouse tumor models. MDOTS profiling demonstrated that TBK1/IKKε inhibition enhanced response to PD-1 blockade, which effectively predicted tumor response in vivo Systematic profiling of secreted cytokines in PDOTS captured key features associated with response and resistance to PD-1 blockade. Thus, MDOTS/PDOTS profiling represents a novel platform to evaluate ICB using established murine models as well as clinically relevant patient specimens.Significance: Resistance to PD-1 blockade remains a challenge for many patients, and biomarkers to guide treatment are lacking. Here, we demonstrate feasibility of ex vivo profiling of PD-1 blockade to interrogate the tumor immune microenvironment, develop therapeutic combinations, and facilitate precision immuno-oncology efforts. Cancer Discov; 8(2); 196-215. ©2017 AACR.See related commentary by Balko and Sosman, p. 143See related article by Deng et al., p. 216This article is highlighted in the In This Issue feature, p. 127.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Citocinas/metabolismo , Resistencia a Antineoplásicos , Citometría de Flujo , Humanos , Inmunohistoquímica , Inmunofenotipificación , Ratones , Técnicas Analíticas Microfluídicas , Receptor de Muerte Celular Programada 1/metabolismo , Esferoides Celulares , Imagen de Lapso de Tiempo , Células Tumorales Cultivadas
15.
Nat Commun ; 8(1): 2193, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29259186

RESUMEN

Despite continued widespread use, the genomic effects of cisplatin-based chemotherapy and implications for subsequent treatment are incompletely characterized. Here, we analyze whole exome sequencing of matched pre- and post-neoadjuvant cisplatin-based chemotherapy primary bladder tumor samples from 30 muscle-invasive bladder cancer patients. We observe no overall increase in tumor mutational burden post-chemotherapy, though a significant proportion of subclonal mutations are unique to the matched pre- or post-treatment tumor, suggesting chemotherapy-induced and/or spatial heterogeneity. We subsequently identify and validate a novel mutational signature in post-treatment tumors consistent with known characteristics of cisplatin damage and repair. We find that post-treatment tumor heterogeneity predicts worse overall survival, and further observe alterations in cell-cycle and immune checkpoint regulation genes in post-treatment tumors. These results provide insight into the clinical and genomic dynamics of tumor evolution with cisplatin-based chemotherapy, suggest mechanisms of clinical resistance, and inform development of clinically relevant biomarkers and trials of combination therapies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma/tratamiento farmacológico , Evolución Clonal/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Vejiga Urinaria/patología , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/genética , Carcinoma/genética , Carcinoma/mortalidad , Carcinoma/patología , Cisplatino/farmacología , Cisplatino/uso terapéutico , Estudios de Cohortes , Cistectomía , Análisis Mutacional de ADN , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Terapia Neoadyuvante/métodos , Invasividad Neoplásica , Tasa de Supervivencia , Transcriptoma/genética , Resultado del Tratamiento , Vejiga Urinaria/cirugía , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/mortalidad , Neoplasias de la Vejiga Urinaria/cirugía , Secuenciación del Exoma
16.
Nature ; 547(7664): 413-418, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28723893

RESUMEN

Immunotherapy with PD-1 checkpoint blockade is effective in only a minority of patients with cancer, suggesting that additional treatment strategies are needed. Here we use a pooled in vivo genetic screening approach using CRISPR-Cas9 genome editing in transplantable tumours in mice treated with immunotherapy to discover previously undescribed immunotherapy targets. We tested 2,368 genes expressed by melanoma cells to identify those that synergize with or cause resistance to checkpoint blockade. We recovered the known immune evasion molecules PD-L1 and CD47, and confirmed that defects in interferon-γ signalling caused resistance to immunotherapy. Tumours were sensitized to immunotherapy by deletion of genes involved in several diverse pathways, including NF-κB signalling, antigen presentation and the unfolded protein response. In addition, deletion of the protein tyrosine phosphatase PTPN2 in tumour cells increased the efficacy of immunotherapy by enhancing interferon-γ-mediated effects on antigen presentation and growth suppression. In vivo genetic screens in tumour models can identify new immunotherapy targets in unanticipated pathways.


Asunto(s)
Sistemas CRISPR-Cas/genética , Edición Génica , Inmunoterapia/métodos , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Escape del Tumor/efectos de los fármacos , Escape del Tumor/inmunología , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Genómica , Humanos , Interferones/inmunología , Mutación con Pérdida de Función , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones , FN-kappa B/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/deficiencia , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Escape del Tumor/genética , Respuesta de Proteína Desplegada , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Immunity ; 46(2): 197-204, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28228279

RESUMEN

Response to immune checkpoint blockade in mesenchymal tumors is poorly characterized, but immunogenomic dissection of these cancers could inform immunotherapy mediators. We identified a treatment-naive patient who has metastatic uterine leiomyosarcoma and has experienced complete tumor remission for >2 years on anti-PD-1 (pembrolizumab) monotherapy. We analyzed the primary tumor, the sole treatment-resistant metastasis, and germline tissue to explore mechanisms of immunotherapy sensitivity and resistance. Both tumors stained diffusely for PD-L2 and showed sparse PD-L1 staining. PD-1+ cell infiltration significantly decreased in the resistant tumor (p = 0.039). Genomically, the treatment-resistant tumor uniquely harbored biallelic PTEN loss and had reduced expression of two neoantigens that demonstrated strong immunoreactivity with patient T cells in vitro, suggesting long-lasting immunological memory. In this near-complete response to PD-1 blockade in a mesenchymal tumor, we identified PTEN mutations and reduced expression of genes encoding neoantigens as potential mediators of resistance to immune checkpoint therapy.


Asunto(s)
Resistencia a Antineoplásicos/genética , Leiomiosarcoma/patología , Fosfohidrolasa PTEN/genética , Neoplasias Uterinas/patología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Análisis Mutacional de ADN , Femenino , Perfilación de la Expresión Génica , Humanos , Leiomiosarcoma/tratamiento farmacológico , Leiomiosarcoma/genética , Persona de Mediana Edad , Mutación , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Transcriptoma , Neoplasias Uterinas/tratamiento farmacológico , Neoplasias Uterinas/genética
19.
Clin Cancer Res ; 23(12): 3214-3222, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-27852700

RESUMEN

Purpose: Squamous cell carcinoma of the anal canal (ASCC) accounts for 2% to 4% of gastrointestinal malignancies in the United States and is increasing in incidence; however, genomic features of ASCC are incompletely characterized. Primary treatment of ASCC involves concurrent chemotherapy and radiation (CRT), but the mutational landscape of resistance to CRT is unknown. Here, we aim to compare mutational features of ASCC in the pre- and post-CRT setting.Experimental Design: We perform whole-exome sequencing of primary (n = 31) and recurrent (n = 30) ASCCs and correlate findings with clinical data. We compare genomic features of matched pre- and post-CRT tumors to identify genomic features of CRT response. Finally, we investigate the mutational underpinnings of an extraordinary ASCC response to immunotherapy.Results: We find that both primary and recurrent ASCC tumors harbor mutations in genes, such as PIK3CA and FBXW7, that are also mutated in other HPV-associated cancers. Overall mutational burden was not significantly different in pre- versus post-CRT tumors, and several examples of shared clonal driver mutations were identified. In two cases, clonally related pre- and post-CRT tumors harbored distinct oncogenic driver mutations in the same cancer gene (KRAS or FBXW7). A patient with recurrent disease achieved an exceptional response to anti-programmed death (PD-1) therapy, and genomic dissection revealed high mutational burden and predicted neoantigen load.Conclusions: We perform comprehensive mutational analysis of ASCC and characterize mutational features associated with CRT. Although many primary and recurrent tumors share driver events, we identify several unique examples of clonal evolution in response to treatment. Clin Cancer Res; 23(12); 3214-22. ©2016 AACR.


Asunto(s)
Neoplasias del Ano/genética , Carcinoma de Células Escamosas/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias del Ano/tratamiento farmacológico , Neoplasias del Ano/patología , Neoplasias del Ano/radioterapia , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/radioterapia , Quimioradioterapia/efectos adversos , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos/genética , Evolución Molecular , Femenino , Genoma Humano/efectos de los fármacos , Genoma Humano/efectos de la radiación , Humanos , Masculino , Persona de Mediana Edad , Mutación/efectos de los fármacos , Mutación/efectos de la radiación , Recurrencia Local de Neoplasia , Tolerancia a Radiación/genética
20.
Cancer Immunol Res ; 5(1): 84-91, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27956380

RESUMEN

Immune checkpoint inhibitors are promising treatments for patients with a variety of malignancies. Toward understanding the determinants of response to immune checkpoint inhibitors, it was previously demonstrated that the presence of somatic mutations is associated with benefit from checkpoint inhibition. A hypothesis was posited that neoantigen homology to pathogens may in part explain the link between somatic mutations and response. To further examine this hypothesis, we reanalyzed cancer exome data obtained from our previously published study of 64 melanoma patients treated with CTLA-4 blockade and a new dataset of RNA-Seq data from 24 of these patients. We found that the ability to accurately predict patient benefit did not increase as the analysis narrowed from somatic mutation burden, to inclusion of only those mutations predicted to be MHC class I neoantigens, to only including those neoantigens that were expressed or that had homology to pathogens. The only association between somatic mutation burden and response was found when examining samples obtained prior to treatment. Neoantigen and expressed neoantigen burden were also associated with response, but neither was more predictive than somatic mutation burden. Neither the previously described tetrapeptide signature nor an updated method to evaluate neoepitope homology to pathogens was more predictive than mutation burden. Cancer Immunol Res; 5(1); 84-91. ©2016 AACR.


Asunto(s)
Antígenos de Neoplasias/genética , Antígeno CTLA-4/antagonistas & inhibidores , Epítopos/genética , Epítopos/inmunología , Melanoma/genética , Melanoma/inmunología , Mutación , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Antígenos de Neoplasias/inmunología , Antineoplásicos Inmunológicos/uso terapéutico , Epítopos/química , Femenino , Humanos , Masculino , Melanoma/tratamiento farmacológico , Melanoma/mortalidad , Persona de Mediana Edad , Pronóstico , Homología de Secuencia de Aminoácido , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...