Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Nature ; 606(7913): 389-395, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35589842

RESUMEN

Cancer immunoediting1 is a hallmark of cancer2 that predicts that lymphocytes kill more immunogenic cancer cells to cause less immunogenic clones to dominate a population. Although proven in mice1,3, whether immunoediting occurs naturally in human cancers remains unclear. Here, to address this, we investigate how 70 human pancreatic cancers evolved over 10 years. We find that, despite having more time to accumulate mutations, rare long-term survivors of pancreatic cancer who have stronger T cell activity in primary tumours develop genetically less heterogeneous recurrent tumours with fewer immunogenic mutations (neoantigens). To quantify whether immunoediting underlies these observations, we infer that a neoantigen is immunogenic (high-quality) by two features-'non-selfness'  based on neoantigen similarity to known antigens4,5, and 'selfness'  based on the antigenic distance required for a neoantigen to differentially bind to the MHC or activate a T cell compared with its wild-type peptide. Using these features, we estimate cancer clone fitness as the aggregate cost of T cells recognizing high-quality neoantigens offset by gains from oncogenic mutations. With this model, we predict the clonal evolution of tumours to reveal that long-term survivors of pancreatic cancer develop recurrent tumours with fewer high-quality neoantigens. Thus, we submit evidence that that the human immune system naturally edits neoantigens. Furthermore, we present a model to predict how immune pressure induces cancer cell populations to evolve over time. More broadly, our results argue that the immune system fundamentally surveils host genetic changes to suppress cancer.


Asunto(s)
Antígenos de Neoplasias , Supervivientes de Cáncer , Neoplasias Pancreáticas , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Linfocitos T/inmunología , Escape del Tumor/inmunología
2.
Nature ; 561(7722): 201-205, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30177826

RESUMEN

Most adult carcinomas develop from noninvasive precursor lesions, a progression that is supported by genetic analysis. However, the evolutionary and genetic relationships among co-existing lesions are unclear. Here we analysed the somatic variants of pancreatic cancers and precursor lesions sampled from distinct regions of the same pancreas. After inferring evolutionary relationships, we found that the ancestral cell had initiated and clonally expanded to form one or more lesions, and that subsequent driver gene mutations eventually led to invasive pancreatic cancer. We estimate that this multi-step progression generally spans many years. These new data reframe the step-wise progression model of pancreatic cancer by illustrating that independent, high-grade pancreatic precursor lesions observed in a single pancreas often represent a single neoplasm that has colonized the ductal system, accumulating spatial and genetic divergence over time.


Asunto(s)
Conductos Pancreáticos/patología , Lesiones Precancerosas/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Linaje de la Célula/genética , Progresión de la Enfermedad , Evolución Molecular , Humanos , Mutación INDEL/genética , Modelos Biológicos , Mutagénesis , Invasividad Neoplásica , Conductos Pancreáticos/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Polimorfismo de Nucleótido Simple/genética , Lesiones Precancerosas/genética , Factores de Tiempo , Secuenciación del Exoma
3.
Genome Res ; 25(10): 1536-45, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26260970

RESUMEN

Somatic L1 retrotransposition events have been shown to occur in epithelial cancers. Here, we attempted to determine how early somatic L1 insertions occurred during the development of gastrointestinal (GI) cancers. Using L1-targeted resequencing (L1-seq), we studied different stages of four colorectal cancers arising from colonic polyps, seven pancreatic carcinomas, as well as seven gastric cancers. Surprisingly, we found somatic L1 insertions not only in all cancer types and metastases but also in colonic adenomas, well-known cancer precursors. Some insertions were also present in low quantities in normal GI tissues, occasionally caught in the act of being clonally fixed in the adjacent tumors. Insertions in adenomas and cancers numbered in the hundreds, and many were present in multiple tumor sections, implying clonal distribution. Our results demonstrate that extensive somatic insertional mutagenesis occurs very early during the development of GI tumors, probably before dysplastic growth.


Asunto(s)
Neoplasias Gastrointestinales/genética , Elementos de Nucleótido Esparcido Largo , Mutagénesis Insercional , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Tiempo
4.
Genes Chromosomes Cancer ; 54(8): 472-481, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26031834

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is driven by the inactivation of the tumor suppressor genes (TSGs), CDKN2A (P16) and SMAD4 (DPC4), commonly by homozygous deletions (HDs). Using a combination of high density single-nucleotide polymorphism (SNP) microarray and whole genome sequencing (WGS), we fine-mapped novel breakpoints surrounding deletions of CDKN2A and SMAD4 and characterized them by their underlying structural variants (SVs). Only one third of CDKN2A and SMAD4 deletions (6 of 18) were simple interstitial deletions, rather, the majority of deletions were caused by complex rearrangements, specifically, a translocation on one side of the TSG in combination with an inversion on the other side. We designate these as "TransFlip" mutations. Characteristics of TransFlip mutations are: (1) a propensity to target the TSGs CDKN2A and SMAD4 (P < 0.005), (2) not present in the germline of the examined samples, (3) non-recurrent breakpoints, (4) relatively small (47 bp to 3.4 kb) inversions, (5) inversions can be either telomeric or centromeric to the TSG, and (6) non-reciprocal, and non-recurrent translocations. TransFlip mutations are novel complex genomic rearrangements with unique breakpoint signatures in pancreatic cancer. We hypothesize that they are a common but poorly understood mechanism of TSG inactivation in human cancer. © 2015 Wiley Periodicals, Inc.

5.
Trends Cancer ; 2024 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-38977383

RESUMEN

Pancreatic cancer is one of the most lethal malignancies, yet much remains to be learned regarding how its precursors develop. In a recent Nature publication, Braxton and Kiemen et al. found that the normal, adult pancreas harbors hundreds to thousands of pancreatic cancer precursors evolving by a variety of routes.

6.
Nat Commun ; 14(1): 749, 2023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-36765116

RESUMEN

Despite insights gained by bulk DNA sequencing of cancer it remains challenging to resolve the admixture of normal and tumor cells, and/or of distinct tumor subclones; high-throughput single-cell DNA sequencing circumvents these and brings cancer genomic studies to higher resolution. However, its application has been limited to liquid tumors or a small batch of solid tumors, mainly because of the lack of a scalable workflow to process solid tumor samples. Here we optimize a highly automated nuclei extraction workflow that achieves fast and reliable targeted single-nucleus DNA library preparation of 38 samples from 16 pancreatic ductal adenocarcinoma patients, with an average library yield per sample of 2867 single nuclei. We demonstrate that this workflow not only performs well using low cellularity or low tumor purity samples but reveals genomic evolution patterns of pancreatic ductal adenocarcinoma as well.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Análisis de Secuencia de ADN , Biblioteca de Genes , Secuenciación de Nucleótidos de Alto Rendimiento
7.
bioRxiv ; 2023 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-36711976

RESUMEN

Multiple large-scale tumor genomic profiling efforts have been undertaken in osteosarcoma, however, little is known about the spatial and temporal intratumor heterogeneity and how it may drive treatment resistance. We performed whole-genome sequencing of 37 tumor samples from eight patients with relapsed or refractory osteosarcoma. Each patient had at least one sample from a primary site and a metastatic or relapse site. We identified subclonal copy number alterations in all but one patient. We observed that in five patients, a subclonal copy number clone from the primary tumor emerged and dominated at subsequent relapses. MYC gain/amplification was enriched in the treatment-resistant clone in 6 out of 7 patients with more than one clone. Amplifications in other potential driver genes, such as CCNE1, RAD21, VEGFA, and IGF1R, were also observed in the resistant copy number clones. Our study sheds light on intratumor heterogeneity and the potential drivers of treatment resistance in osteosarcoma.

8.
Cancer Res ; 83(22): 3796-3812, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37812025

RESUMEN

Multiple large-scale genomic profiling efforts have been undertaken in osteosarcoma to define the genomic drivers of tumorigenesis, therapeutic response, and disease recurrence. The spatial and temporal intratumor heterogeneity could also play a role in promoting tumor growth and treatment resistance. We conducted longitudinal whole-genome sequencing of 37 tumor samples from 8 patients with relapsed or refractory osteosarcoma. Each patient had at least one sample from a primary site and a metastatic or relapse site. Subclonal copy-number alterations were identified in all patients except one. In 5 patients, subclones from the primary tumor emerged and dominated at subsequent relapses. MYC gain/amplification was enriched in the treatment-resistant clones in 6 of 7 patients with multiple clones. Amplifications in other potential driver genes, such as CCNE1, RAD21, VEGFA, and IGF1R, were also observed in the resistant copy-number clones. A chromosomal duplication timing analysis revealed that complex genomic rearrangements typically occurred prior to diagnosis, supporting a macroevolutionary model of evolution, where a large number of genomic aberrations are acquired over a short period of time followed by clonal selection, as opposed to ongoing evolution. A mutational signature analysis of recurrent tumors revealed that homologous repair deficiency (HRD)-related SBS3 increases at each time point in patients with recurrent disease, suggesting that HRD continues to be an active mutagenic process after diagnosis. Overall, by examining the clonal relationships between temporally and spatially separated samples from patients with relapsed/refractory osteosarcoma, this study sheds light on the intratumor heterogeneity and potential drivers of treatment resistance in this disease. SIGNIFICANCE: The chemoresistant population in recurrent osteosarcoma is subclonal at diagnosis, emerges at the time of primary resection due to selective pressure from neoadjuvant chemotherapy, and is characterized by unique oncogenic amplifications.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Humanos , Osteosarcoma/genética , Secuenciación Completa del Genoma , Genómica , Neoplasias Óseas/genética , Recurrencia , Variaciones en el Número de Copia de ADN , Mutación
9.
Cell Rep ; 39(5): 110771, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35508134

RESUMEN

We performed a comparative analysis of human and 12 non-human primates to identify sequence variations in known cancer genes. We identified 395 human-specific fixed non-silent substitutions that emerged during evolution of human. Using bioinformatics analyses for functional consequences, we identified a number of substitutions that are predicted to alter protein function; one of these mutations is located at the most evolutionarily conserved domain of human BRCA2.


Asunto(s)
Pan troglodytes , Primates , Animales , Proteína BRCA2/genética , Evolución Molecular , Humanos , Mutación/genética , Pan troglodytes/genética , Proteínas/metabolismo
10.
Clin Cancer Res ; 27(5): 1516-1525, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33323400

RESUMEN

PURPOSE: Melanoma is a biologically heterogeneous disease composed of distinct clinicopathologic subtypes that frequently resist treatment. To explore the evolution of treatment resistance and metastasis, we used a combination of temporal and multilesional tumor sampling in conjunction with whole-exome sequencing of 110 tumors collected from 7 patients with cutaneous (n = 3), uveal (n = 2), and acral (n = 2) melanoma subtypes. EXPERIMENTAL DESIGN: Primary tumors, metastases collected longitudinally, and autopsy tissues were interrogated. All but 1 patient died because of melanoma progression. RESULTS: For each patient, we generated phylogenies and quantified the extent of genetic diversity among tumors, specifically among putative somatic alterations affecting therapeutic resistance. CONCLUSIONS: In 4 patients who received immunotherapy, we found 1-3 putative acquired and intrinsic resistance mechanisms coexisting in the same patient, including mechanisms that were shared by all tumors within each patient, suggesting that future therapies directed at overcoming intrinsic resistance mechanisms may be broadly effective.


Asunto(s)
Resistencia a Antineoplásicos/genética , Evolución Molecular , Inmunoterapia/métodos , Melanoma/patología , Mutación , Neoplasias Cutáneas/patología , Neoplasias de la Úvea/patología , Biomarcadores de Tumor , Humanos , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/inmunología , Pronóstico , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias de la Úvea/tratamiento farmacológico , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/inmunología
11.
Nat Cancer ; 1(1): 59-74, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-35118421

RESUMEN

Pancreatic cancer expression profiles largely reflect a classical or basal-like phenotype. The extent to which these profiles vary within a patient is unknown. We integrated evolutionary analysis and expression profiling in multiregion-sampled metastatic pancreatic cancers, finding that squamous features are the histologic correlate of an RNA-seq-defined basal-like subtype. In patients with coexisting basal and squamous and classical and glandular morphology, phylogenetic studies revealed that squamous morphology represented a subclonal population in an otherwise classical and glandular tumor. Cancers with squamous features were significantly more likely to have clonal mutations in chromatin modifiers, intercellular heterogeneity for MYC amplification and entosis. These data provide a unifying paradigm for integrating basal-type expression profiles, squamous histology and somatic mutations in chromatin modifier genes in the context of clonal evolution of pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático , Carcinoma de Células Escamosas , Neoplasias Pancreáticas , Carcinoma Ductal Pancreático/genética , Carcinoma de Células Escamosas/genética , Cromatina , Humanos , Neoplasias Pancreáticas/genética , Filogenia , Neoplasias Pancreáticas
12.
Cancer Discov ; 10(6): 792-805, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32193223

RESUMEN

Surgery is the only curative option for stage I/II pancreatic cancer; nonetheless, most patients will experience a recurrence after surgery and die of their disease. To identify novel opportunities for management of recurrent pancreatic cancer, we performed whole-exome or targeted sequencing of 10 resected primary cancers and matched intrapancreatic recurrences or distant metastases. We identified that recurrent disease after adjuvant or first-line platinum therapy corresponds to an increased mutational burden. Recurrent disease is enriched for genetic alterations predicted to activate MAPK/ERK and PI3K-AKT signaling and develops from a monophyletic or polyphyletic origin. Treatment-induced genetic bottlenecks lead to a modified genetic landscape and subclonal heterogeneity for driver gene alterations in part due to intermetastatic seeding. In 1 patient what was believed to be recurrent disease was an independent (second) primary tumor. These findings suggest routine post-treatment sampling may have value in the management of recurrent pancreatic cancer. SIGNIFICANCE: The biological features or clinical vulnerabilities of recurrent pancreatic cancer after pancreaticoduodenectomy are unknown. Using whole-exome sequencing we find that recurrent disease has a distinct genomic landscape, intermetastatic genetic heterogeneity, diverse clonal origins, and higher mutational burden than found for treatment-naïve disease.See related commentary by Bednar and Pasca di Magliano, p. 762.This article is highlighted in the In This Issue feature, p. 747.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Metástasis de la Neoplasia/genética , Recurrencia Local de Neoplasia/genética , Neoplasias Pancreáticas/genética , Carcinoma Ductal Pancreático/secundario , Evolución Molecular , Humanos , Recurrencia Local de Neoplasia/patología , Neoplasias Pancreáticas/patología , Secuenciación del Exoma
13.
Nat Commun ; 11(1): 3617, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32680998

RESUMEN

Multiple myeloma (MM) progression is characterized by the seeding of cancer cells in different anatomic sites. To characterize this evolutionary process, we interrogated, by whole genome sequencing, 25 samples collected at autopsy from 4 patients with relapsed MM and an additional set of 125 whole exomes collected from 51 patients. Mutational signatures analysis showed how cytotoxic agents introduce hundreds of unique mutations in each surviving cancer cell, detectable by bulk sequencing only in cases of clonal expansion of a single cancer cell bearing the mutational signature. Thus, a unique, single-cell genomic barcode can link chemotherapy exposure to a discrete time window in a patient's life. We leveraged this concept to show that MM systemic seeding is accelerated at relapse and appears to be driven by the survival and subsequent expansion of a single myeloma cell following treatment with high-dose melphalan therapy and autologous stem cell transplant.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Evolución Clonal/efectos de los fármacos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia/patología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Melfalán/administración & dosificación , Melfalán/efectos adversos , Persona de Mediana Edad , Mieloma Múltiple/diagnóstico por imagen , Mieloma Múltiple/genética , Mieloma Múltiple/terapia , Mutación/efectos de los fármacos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Recurrencia Local de Neoplasia/diagnóstico por imagen , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/terapia , Tomografía Computarizada por Tomografía de Emisión de Positrones , Análisis de la Célula Individual , Análisis Espacio-Temporal , Trasplante Autólogo/efectos adversos , Secuenciación Completa del Genoma
14.
Nat Rev Cancer ; 19(11): 639-650, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31455892

RESUMEN

Genetic intratumoural heterogeneity is a natural consequence of imperfect DNA replication. Any two randomly selected cells, whether normal or cancerous, are therefore genetically different. Here, we review the different forms of genetic heterogeneity in cancer and re-analyse the extent of genetic heterogeneity within seven types of untreated epithelial cancers, with particular regard to its clinical relevance. We find that the homogeneity of predicted functional mutations in driver genes is the rule rather than the exception. In primary tumours with multiple samples, 97% of driver-gene mutations in 38 patients were homogeneous. Moreover, among metastases from the same primary tumour, 100% of the driver mutations in 17 patients were homogeneous. With a single biopsy of a primary tumour in 14 patients, the likelihood of missing a functional driver-gene mutation that was present in all metastases was 2.6%. Furthermore, all functional driver-gene mutations detected in these 14 primary tumours were present among all their metastases. Finally, we found that individual metastatic lesions responded concordantly to targeted therapies in 91% of 44 patients. These analyses indicate that the cells within the primary tumours that gave rise to metastases are genetically homogeneous with respect to functional driver-gene mutations, and we suggest that future efforts to develop combination therapies have the potential to be curative.


Asunto(s)
Heterogeneidad Genética , Mutación , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Animales , Biopsia , Ensayos Clínicos como Asunto , Epigénesis Genética , Humanos , Oncología Médica , Metástasis de la Neoplasia
15.
Nat Commun ; 10(1): 5435, 2019 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-31780749

RESUMEN

The KPC mouse model, driven by the Kras and Trp53 transgenes, is well regarded for faithful recapitulation of human pancreatic cancer biology. However, the extent that this model recapitulates the subclonal evolution of this tumor type is unknown. Here we report evidence of continuing subclonal evolution after tumor initiation that largely reflect copy number alterations that target cellular processes of established significance in human pancreatic cancer. The evolutionary trajectories of the mouse tumors show both linear and branching patterns as well as clonal mixing. We propose the KPC model and derivatives have unexplored utility as a functional system to model the mechanisms and modifiers of tumor evolution.


Asunto(s)
Adenocarcinoma/genética , Neoplasias Pancreáticas/genética , Animales , Evolución Clonal/genética , Variaciones en el Número de Copia de ADN/genética , Modelos Animales de Enfermedad , Evolución Molecular , Ratones , Ratones Transgénicos , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Proteína p53 Supresora de Tumor/genética
17.
JCO Precis Oncol ; 7: e2300170, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37285558
18.
Science ; 361(6406): 1033-1037, 2018 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-30190408

RESUMEN

Metastases are responsible for the majority of cancer-related deaths. Although genomic heterogeneity within primary tumors is associated with relapse, heterogeneity among treatment-naïve metastases has not been comprehensively assessed. We analyzed sequencing data for 76 untreated metastases from 20 patients and inferred cancer phylogenies for breast, colorectal, endometrial, gastric, lung, melanoma, pancreatic, and prostate cancers. We found that within individual patients, a large majority of driver gene mutations are common to all metastases. Further analysis revealed that the driver gene mutations that were not shared by all metastases are unlikely to have functional consequences. A mathematical model of tumor evolution and metastasis formation provides an explanation for the observed driver gene homogeneity. Thus, single biopsies capture most of the functionally important mutations in metastases and therefore provide essential information for therapeutic decision-making.


Asunto(s)
Heterogeneidad Genética , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Humanos , Modelos Teóricos , Mutación , Metástasis de la Neoplasia/patología , Neoplasias/patología
19.
Oncotarget ; 8(26): 41792-41805, 2017 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-28611298

RESUMEN

Cancer is an evolutionary disease, and there is increasing interest in applying tools from evolutionary biology to understand cancer progression. Restriction-site associated DNA sequencing (RADseq) was developed for the field of evolutionary genetics to study adaptation and identify evolutionary relationships among populations. Here we apply RADseq to study tumor evolution, which allows for unbiased sampling of any desired frequency of the genome, overcoming the selection bias and cost limitations inherent to exome or whole-genome sequencing. We apply RADseq to both human pancreatic cancer and zebrafish melanoma samples. Using either a low-frequency (SbfI, 0.4% of the genome) or high-frequency (NsiI, 6-9% of the genome) cutter, we successfully identify single nucleotide substitutions and copy number alterations in tumors, which can be augmented by performing RADseq on sublineages within the tumor. We are able to infer phylogenetic relationships between primary tumors and metastases. These same methods can be used to identify somatic mosaicism in seemingly normal, non-cancerous tissues. Evolutionary studies of cancer that focus on rates of tumor evolution and evolutionary relationships among tumor lineages will benefit from the flexibility and efficiency of restriction-site associated DNA sequencing.


Asunto(s)
Predisposición Genética a la Enfermedad , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Neoplasias/genética , Neoplasias/patología , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Estudios de Asociación Genética , Genómica/métodos , Humanos , Mosaicismo , Clasificación del Tumor , Metástasis de la Neoplasia , Estadificación de Neoplasias , Secuenciación del Exoma , Secuenciación Completa del Genoma , Pez Cebra
20.
Artículo en Inglés | MEDLINE | ID: mdl-28679692

RESUMEN

We describe an 85-yr-old male of Ashkenazi Jewish descent with biopsy-proven locally advanced pancreatic ductal adenocarcinoma (PDA). The patient underwent a modified course of gemcitabine and stereotactic body radiation therapy and survived for 42 mo with a stable pancreatic head mass and no evidence of metastatic disease before death due to complications from a stroke. Whole-exome sequencing of his tumor revealed a simple genome landscape with no evidence of mutations, copy-number changes, or structural alterations in genes most commonly associated with PDA (i.e., KRAS, CDKN2A, TP53, or SMAD4). An analysis of his germline DNA revealed no pathogenic variants of significance. Whole-exome and whole-genome sequencing identified a somatic mutation of RNF213 and an inversion/deletion of CTNNA2 as the genetic basis of his PDA. Although PDA is classically characterized by a predictable set of mutations, these data suggest that alternate genetic paths to PDA may exist, which can be associated with a more indolent clinical course.


Asunto(s)
Adenosina Trifosfatasas/genética , Carcinoma Ductal Pancreático/genética , Ubiquitina-Proteína Ligasas/genética , alfa Catenina/genética , Adenocarcinoma/genética , Adenosina Trifosfatasas/metabolismo , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Exoma , Genómica , Humanos , Mutación INDEL/genética , Mutación , Páncreas/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Secuenciación del Exoma , alfa Catenina/metabolismo , Gemcitabina , Neoplasias Pancreáticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA