Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Nature ; 538(7625): 378-382, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27732578

ABSTRACT

Pancreatic cancer, a highly aggressive tumour type with uniformly poor prognosis, exemplifies the classically held view of stepwise cancer development. The current model of tumorigenesis, based on analyses of precursor lesions, termed pancreatic intraepithelial neoplasm (PanINs) lesions, makes two predictions: first, that pancreatic cancer develops through a particular sequence of genetic alterations (KRAS, followed by CDKN2A, then TP53 and SMAD4); and second, that the evolutionary trajectory of pancreatic cancer progression is gradual because each alteration is acquired independently. A shortcoming of this model is that clonally expanded precursor lesions do not always belong to the tumour lineage, indicating that the evolutionary trajectory of the tumour lineage and precursor lesions can be divergent. This prevailing model of tumorigenesis has contributed to the clinical notion that pancreatic cancer evolves slowly and presents at a late stage. However, the propensity for this disease to rapidly metastasize and the inability to improve patient outcomes, despite efforts aimed at early detection, suggest that pancreatic cancer progression is not gradual. Here, using newly developed informatics tools, we tracked changes in DNA copy number and their associated rearrangements in tumour-enriched genomes and found that pancreatic cancer tumorigenesis is neither gradual nor follows the accepted mutation order. Two-thirds of tumours harbour complex rearrangement patterns associated with mitotic errors, consistent with punctuated equilibrium as the principal evolutionary trajectory. In a subset of cases, the consequence of such errors is the simultaneous, rather than sequential, knockout of canonical preneoplastic genetic drivers that are likely to set-off invasive cancer growth. These findings challenge the current progression model of pancreatic cancer and provide insights into the mutational processes that give rise to these aggressive tumours.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/pathology , Gene Rearrangement/genetics , Genome, Human/genetics , Models, Biological , Mutagenesis/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Carcinoma in Situ/genetics , Chromothripsis , DNA Copy Number Variations/genetics , Disease Progression , Evolution, Molecular , Female , Genes, Neoplasm/genetics , Humans , Male , Mitosis/genetics , Mutation/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Polyploidy , Precancerous Conditions/genetics
2.
PLoS Comput Biol ; 15(1): e1006596, 2019 01.
Article in English | MEDLINE | ID: mdl-30629588

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has the worst prognosis among solid malignancies and improved therapeutic strategies are needed to improve outcomes. Patient-derived xenografts (PDX) and patient-derived organoids (PDO) serve as promising tools to identify new drugs with therapeutic potential in PDAC. For these preclinical disease models to be effective, they should both recapitulate the molecular heterogeneity of PDAC and validate patient-specific therapeutic sensitivities. To date however, deep characterization of the molecular heterogeneity of PDAC PDX and PDO models and comparison with matched human tumour remains largely unaddressed at the whole genome level. We conducted a comprehensive assessment of the genetic landscape of 16 whole-genome pairs of tumours and matched PDX, from primary PDAC and liver metastasis, including a unique cohort of 5 'trios' of matched primary tumour, PDX, and PDO. We developed a pipeline to score concordance between PDAC models and their paired human tumours for genomic events, including mutations, structural variations, and copy number variations. Tumour-model comparisons of mutations displayed single-gene concordance across major PDAC driver genes, but relatively poor agreement across the greater mutational load. Genome-wide and chromosome-centric analysis of structural variation (SV) events highlights previously unrecognized concordance across chromosomes that demonstrate clustered SV events. We found that polyploidy presented a major challenge when assessing copy number changes; however, ploidy-corrected copy number states suggest good agreement between donor-model pairs. Collectively, our investigations highlight that while PDXs and PDOs may serve as tractable and transplantable systems for probing the molecular properties of PDAC, these models may best serve selective analyses across different levels of genomic complexity.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Genome/genetics , Models, Biological , Neoplasms, Experimental/genetics , Pancreatic Neoplasms/genetics , Animals , Biomedical Research/standards , Humans , Pancreas/pathology
4.
Proc Natl Acad Sci U S A ; 108(17): 7160-5, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21474781

ABSTRACT

The tumor microenvironment strongly influences cancer development, progression, and metastasis. The role of carcinoma-associated fibroblasts (CAFs) in these processes and their clinical impact has not been studied systematically in non-small cell lung carcinoma (NSCLC). We established primary cultures of CAFs and matched normal fibroblasts (NFs) from 15 resected NSCLC. We demonstrate that CAFs have greater ability than NFs to enhance the tumorigenicity of lung cancer cell lines. Microarray gene-expression analysis of the 15 matched CAF and NF cell lines identified 46 differentially expressed genes, encoding for proteins that are significantly enriched for extracellular proteins regulated by the TGF-ß signaling pathway. We have identified a subset of 11 genes (13 probe sets) that formed a prognostic gene-expression signature, which was validated in multiple independent NSCLC microarray datasets. Functional annotation using protein-protein interaction analyses of these and published cancer stroma-associated gene-expression changes revealed prominent involvement of the focal adhesion and MAPK signaling pathways. Fourteen (30%) of the 46 genes also were differentially expressed in laser-capture-microdissected corresponding primary tumor stroma compared with the matched normal lung. Six of these 14 genes could be induced by TGF-ß1 in NF. The results establish the prognostic impact of CAF-associated gene-expression changes in NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/metabolism , Neoplasm Proteins/biosynthesis , Animals , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Transformed , Disease-Free Survival , Female , Fibroblasts/pathology , Gene Expression Profiling , Humans , Lung/metabolism , Lung/pathology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mice , Mice, SCID , Oligonucleotide Array Sequence Analysis , Signal Transduction , Survival Rate
5.
Sci Rep ; 11(1): 10619, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34011980

ABSTRACT

Patient-derived xenograft (PDX) and their xenograft-derived organoid (XDO) models that recapitulate the genotypic and phenotypic landscape of patient cancers could help to advance research and lead to improved clinical management. PDX models were established from 276 pancreato-duodenal and biliary cancer resections. Initial, passage 0 (P0) engraftment rates were 59% (118/199) for pancreatic, 86% (25/29) for duodenal, and 35% (17/48) for biliary ductal tumors. Pancreatic ductal adenocarcinoma (PDAC), had a P0 engraftment rate of 62% (105/169). KRAS mutant and wild-type PDAC models were molecularly profiled, and XDO models were generated to perform initial drug response evaluations. Subsets of PDAC PDX models showed global copy number variants and gene expression profiles that were retained with serial passaging, and they showed a spectrum of somatic mutations represented in patient tumors. PDAC XDO models were established, with a success rate of 71% (10/14). Pathway activation of KRAS-MAPK in PDXs was independent of KRAS mutational status. Four wild-type KRAS models were characterized by one with EGFR (L747-P753 del), two with BRAF alterations (N486_P490del or V600E), and one with triple negative KRAS/EGFR/BRAF. Model OCIP256, characterized by BRAF (N486-P490 del), had activated phospho-ERK. A combination treatment of a pan-RAF inhibitor (LY3009120) and a MEK inhibitor (trametinib) effectively suppressed phospho-ERK and inhibited growth of OCIP256 XDO and PDX models. PDAC/duodenal adenocarcinoma have high success rates forming PDX/organoid and retaining their phenotypic and genotypic features. These models may be effective tools to evaluate novel drug combination therapies.


Subject(s)
Biliary Tract Neoplasms/pathology , Duodenal Neoplasms/pathology , Organoids/pathology , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biliary Tract Neoplasms/drug therapy , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Duodenal Neoplasms/drug therapy , Humans , Mice, Inbred NOD , Mice, SCID , Mutation/genetics , Organoids/drug effects , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms
6.
Mol Cancer Res ; 15(1): 3-14, 2017 01.
Article in English | MEDLINE | ID: mdl-27678171

ABSTRACT

Carcinoma-associated fibroblasts (CAF) represent a significant component of pancreatic cancer stroma and are biologically implicated in tumor progression. However, evidence of both cancer-promoting and -restraining properties amongst CAFs suggests the possibility of multiple phenotypic subtypes. Here, it is demonstrated that senescent CAFs promote pancreatic cancer invasion and metastasis compared with nonsenescent control CAFs using in vitro Transwell invasion models and in vivo xenograft mouse models. Screening by gene expression microarray and cytokine ELISA assays revealed IL8 to be upregulated in senescent CAFs. Experimental modulation through IL8 overexpression or receptor inhibition implicates the IL8 pathway as a mediator of the proinvasive effects of senescent CAFs. In a cohort of human pancreatic cancer cases, more abundant stromal senescence as indicated by p16 immunohistochemistry correlated with decreased survival in patients with early-stage disease. These data support senescent fibroblasts as a pathologically and clinically relevant feature of pancreatic cancer. The inhibition of senescent stroma-cancer signaling pathways has the potential to restrain pancreatic cancer progression. IMPLICATIONS: Findings show that senescent cancer-associated fibroblasts secret excess IL8 to promote pancreatic cancer invasion and metastasis; thus, senescent CAFs represent a phenotypic subtype, challenging conventional assumptions that CAFs are a homogeneous population. Mol Cancer Res; 15(1); 3-14. ©2016 AACR.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Cellular Senescence , Interleukin-8/metabolism , Pancreatic Neoplasms/pathology , Up-Regulation , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Male , Mice , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Phenotype , Stromal Cells/pathology
7.
Cancers (Basel) ; 6(1): 459-71, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24577243

ABSTRACT

The unusually dense stroma of pancreatic cancers is thought to play an important role in their biological aggression. The presence of hypoxia is also considered an adverse prognostic factor. Although it is usually assumed that this is the result of effects of hypoxia on the epithelial component, it is possible that hypoxia exerts indirect effects via the tumor stroma. We therefore measured hypoxia in the stroma of a series of primary pancreatic cancer xenografts. Nine patient-derived pancreatic xenografts representing a range of oxygenation levels were labeled by immunohistochemistry for EF5 and analyzed using semi-automated pattern recognition software. Hypoxia in the tumor and stroma was correlated with tumor growth and metastatic potential. The extent of hypoxia varied from 1%-39% between the different models. EF5 labeling in the stroma ranged from 0-20% between models, and was correlated with the level of hypoxia in the tumor cell area, but not microvessel density. Tumor hypoxia correlated with spontaneous metastasis formation with the exception of one hypoxic model that showed disproportionately low levels of hypoxia in the stroma and was non-metastatic. Our results demonstrate that hypoxia exists in the stroma of primary pancreatic cancer xenografts and suggest that stromal hypoxia impacts the metastatic potential.

8.
PLoS One ; 8(12): e84366, 2013.
Article in English | MEDLINE | ID: mdl-24386371

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is the fourth most common cause of cancer death in North America. Activating KRAS mutations and Smad4 loss occur in approximately 90% and 55% of PDAC, respectively. While their roles in the early stages of PDAC development have been confirmed in genetically modified mouse models, their roles in the multistep malignant transformation of human pancreatic duct cells have not been directly demonstrated. Here, we report that Smad4 represents a barrier in KRAS-mediated malignant transformation of the near normal immortalized human pancreatic duct epithelial (HPDE) cell line model. Marked Smad4 downregulation by shRNA in KRAS (G12V) expressing HPDE cells failed to cause tumorigenic transformation. However, KRAS-mediated malignant transformation occurred in a new HPDE-TGF-ß resistant (TßR) cell line that completely lacks Smad4 protein expression and is resistant to the mito-inhibitory activity of TGF-ß. This transformation resulted in tumor formation and development of metastatic phenotype when the cells were implanted orthotopically into the mouse pancreas. Smad4 restoration re-established TGF-ß sensitivity, markedly increased tumor latency by promoting apoptosis, and decreased metastatic potential. These results directly establish the critical combination of the KRAS oncogene and complete Smad4 inactivation in the multi-stage malignant transformation and metastatic progression of normal human HPDE cells.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic , Epithelial Cells/pathology , Pancreatic Ducts/pathology , Proto-Oncogene Proteins/metabolism , Signal Transduction , Smad4 Protein/metabolism , ras Proteins/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Gene Silencing , Humans , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Proto-Oncogene Proteins p21(ras) , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Smad4 Protein/deficiency , Smad4 Protein/genetics , Survival Analysis , Transforming Growth Factor beta/pharmacology
9.
Clin Cancer Res ; 19(1): 118-27, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23091117

ABSTRACT

PURPOSE: MEK inhibition has clinical activity against biliary cancers and might therefore be successfully combined with gemcitabine, one of the most active chemotherapy agents for these cancers. As gemcitabine is active in S-phase, and the extracellular signal-regulated kinase (ERK) pathway has a major role driving cell-cycle progression, concurrent use of a MEK inhibitor could potentially antagonize the effect of gemcitabine. We therefore tested the sequence dependence of the combination of gemcitabine and the MEK inhibitor AZD6244 using a series of biliary cancer models. EXPERIMENTAL DESIGN: Primary xenografts were established from patients with gallbladder and distal bile duct cancer and grown in severe combined immunodeficient (SCID) mice at the subcutaneous site. Plasma and tumor drug levels and the time course for recovery of ERK signaling and S-phase were measured in tumor-bearing mice treated for 48 hours with AZD6244 and then monitored for 48 hours off treatment. On the basis of these results, two different treatment schedules combining AZD6244 with gemcitabine were tested in four different biliary cancer models. RESULTS: DNA synthesis was suppressed during treatment with AZD6244, and reentry into S-phase was delayed by approximately 48 hours after treatment. Strong schedule dependence was seen in all four biliary cancer models tested, suggesting that combined treatment with AZD6244 plus gemcitabine would be more active in patients with biliary cancer when gemcitabine is given following a 48-hour interruption in AZD6244 dosing, rather than concurrently. CONCLUSIONS: The combination of AZD6244 plus gemcitabine is highly schedule dependent, and predicted to be more effective in the clinic using sequential rather than simultaneous dosing protocols.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/pharmacology , Biliary Tract Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacokinetics , Biliary Tract Neoplasms/enzymology , Biliary Tract Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Disease Models, Animal , Drug Therapy, Combination , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Male , Mice , Mice, SCID , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
10.
Neoplasia ; 11(12): 1292-300, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20019837

ABSTRACT

Complete resection of early-stage non-small cell lung cancer (NSCLC) is potentially curative, yet approximately 50% of patients are at risk for developing metastatic recurrence. Met, the receptor for hepatocyte growth factor (HGF) is a receptor tyrosine kinase with demonstrated roles in regulating cellular proliferation, motility, morphogenesis, and apoptosis. Met receptor and its ligand, HGF, are commonly overexpressed in NSCLC, and their overexpression has been associated with poor prognosis, which could potentially involve a paracrine and/or autocrine activation loop. However, there is as yet no direct evidence that HGF-Met signaling directly promotes metastasis in NSCLC cells. Using retroviral transduction, we overexpressed the human c-met and hgf complementary DNA, alone or in combination in the NCI-H460 human large cell carcinoma cell line. The HGF/Met co-overexpressing (H460-HGF/Met) cells demonstrated enhanced tumorigenicity in xenograft SCID mice. When these cells are implanted orthotopically into the lungs of nude rats, only the H460-HGF/Met cells showed higher spontaneous metastases to distant organs including bone, brain, and kidney. These results provide evidence that autocrine overactivation of the Met- HGF loop enhances systemic metastases in NSCLC. Targeted interference of this loop may potentially be an effective adjuvant therapy to improve survival of early-stage NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Hepatocyte Growth Factor/metabolism , Lung Neoplasms/pathology , Proto-Oncogene Proteins c-met/metabolism , Receptors, Growth Factor/metabolism , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Carcinoma, Large Cell/genetics , Carcinoma, Large Cell/metabolism , Carcinoma, Large Cell/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/genetics , Humans , Immunohistochemistry , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/secondary , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, SCID , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-met/genetics , Rats , Rats, Nude , Receptors, Growth Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL