Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Philos Ethics Humanit Med ; 18(1): 5, 2023 05 24.
Article in English | MEDLINE | ID: mdl-37221540

ABSTRACT

BACKGROUND: Biomedicine, i.e. the application of basic sciences to medicine, has become the cornerstone for the study of etiopathogenesis and treatment of diseases. Biomedicine has enormously contributed to the progress of medicine and healthcare and has become the preferred approach to medical problems in the West. The developments in statistical inference and machine learning techniques have provided the foundation for personalised medicine where clinical management can be fully informed by biomedicine. The deployment of precision medicine may impact the autonomy and self-normativity of the patients. Understanding the relationship between biomedicine and medical practice can help navigate the benefits and challenges offered by precision medicine. METHODS: Conventional content analysis was applied to "Le Normal and le Pathologique" (Canguilhem G. The Normal and the Pathological. Princeton: Princeton University Press; 1991) and further investigated with respect to its relationship with techne and precision medicine using PubMed and Google Scholar and the Standford Encyclopedia of Philosophy to search for the following keywords singularly or in combination: "Canguilhem", "techne", "episteme", "precision medicine", "machine learning AND medicine". RESULTS: The Hippocratic concept of techne accounts for many characteristics of medical knowledge and practice. The advances of biomedicine, experimental medicine and, more recently, machine learning offer, in contrast, the model of a medicine based purely on episteme. I argue that Canguilhem medical epistemology establishes a framework where episteme and data-driven medicine is compatible with the promotion of patient's autonomy and self-normativity. CONCLUSIONS: Canguilhem's medical epistemology orders the relationship of applied medicine with experimental sciences, ethics and social sciences. It provides guidance to define the scope of medicine and the boundaries of medicalization of healthy life. Finally, it sets an agenda for a safe implementation of machine learning in medicine.


Subject(s)
Biomedical Research , Lepidoptera , Humans , Animals , Precision Medicine , Health Occupations , Health Facilities , Health Status
3.
Commun Biol ; 5(1): 335, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35396535

ABSTRACT

Oesophageal adenocarcinoma (OAC) provides an ideal case study to characterize large-scale rearrangements. Using whole genome short-read sequencing of 383 cases, for which 214 had matched whole transcriptomes, we observed structural variations (SV) with a predominance of deletions, tandem duplications and inter-chromosome junctions that could be identified as LINE-1 mobile element (ME) insertions. Complex clusters of rearrangements resembling breakage-fusion-bridge cycles or extrachromosomal circular DNA accounted for 22% of complex SVs affecting known oncogenes. Counting SV events affecting known driver genes substantially increased the recurrence rates of these drivers. After excluding fragile sites, we identified 51 candidate new drivers in genomic regions disrupted by SVs, including ETV5, KAT6B and CLTC. RUNX1 was the most recurrently altered gene (24%), with many deletions inactivating the RUNT domain but preserved the reading frame, suggesting an altered protein product. These findings underscore the importance of identification of SV events in OAC with implications for targeted therapies.


Subject(s)
Adenocarcinoma , Esophageal Neoplasms , Adenocarcinoma/genetics , Esophageal Neoplasms/genetics , Genome, Human , Histone Acetyltransferases/genetics , Humans , Whole Genome Sequencing
4.
EClinicalMedicine ; 42: 101183, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34805809

ABSTRACT

BACKGROUND: Neoadjuvant chemoradiotherapy (NCRT) or neoadjuvant chemotherapy (NCT) followed by surgery are two standard strategies in treating locally advanced esophageal cancer (EC). We aim to compare NCRT and NCT in the management of locally advanced EC patients. METHODS: MEDLINE, Embase, CENTRAL, and conferences were systematically searched for clinical trials published up to September 2021. Pairwise comparisons and Bayesian network meta-analyses were conducted to compare overall survival (OS) and disease-free survival (DFS) by reporting the hazard ratio (HR) and 95% credible intervals (CrIs). The study was registered at PROSPERO (CRD42020170619). FINDINGS: 25 trials with 4563 EC patients met inclusion criteria. NCRT improved OS (HR: 0·72, 95%CrI: 0·63-0·82) and DFS (HR: 0·72, 95%CrI: 0·63-0·81) compared to surgery alone. NCRT improved OS (HR: 0·83, 95%CrI: 0·69-0·99) and DFS (HR: 0·83, 95%CI: 0·69-0·99) compared to NCT. Subgroup analysis demonstrated that both NCRT (HR: 0·77, 95%CrI: 0·65-0·90) and NCT (HR: 0·81, 95%CrI: 0·67-0·99) improved OS than surgery in esophageal squamous cell carcinoma (ESCC) patients. No significant differences were observed between NCRT and NCT regarding OS (HR: 0·95, 95%CrI: 0·75-1·19) and DFS (HR: 0·90, 95%CrI: 0·50-1·62) in ESCC. The short-term outcomes were similar between NCRT and NCT. The three treatment strategies were comparable in esophageal adenocarcinoma (EAC) subpopulations. INTERPRETATION: The study corroborated current guidelines in addressing the importance of analysing EC according to histopathological types. The analysis suggested that in locally advanced ESCC patients, both NCRT and NCT improved OS as compared to surgery alone, whereas no clear evidence supported the optimal strategies between NCRT and NCT. More RCTs comparing different therapeutic strategies in EAC patients are warranted. FUNDING: Köln Fortune Program, University of Cologne.

5.
Cancers (Basel) ; 13(11)2021 Jun 03.
Article in English | MEDLINE | ID: mdl-34205004

ABSTRACT

Sporadic cancer develops from the accrual of somatic mutations. Out of all small-scale somatic aberrations in coding regions, 95% are base substitutions, with 90% being missense mutations. While multiple studies focused on the importance of this mutation type, a machine learning method based on the number of protein-protein interactions (PPIs) has not been fully explored. This study aims to develop an improved computational method for driver identification, validation and evaluation (DRIVE), which is compared to other methods for assessing its performance. DRIVE aims at distinguishing between driver and passenger mutations using a feature-based learning approach comprising two levels of biological classification for a pan-cancer assessment of somatic mutations. Gene-level features include the maximum number of protein-protein interactions, the biological process and the type of post-translational modifications (PTMs) while mutation-level features are based on pathogenicity scores. Multiple supervised classification algorithms were trained on Genomics Evidence Neoplasia Information Exchange (GENIE) project data and then tested on an independent dataset from The Cancer Genome Atlas (TCGA) study. Finally, the most powerful classifier using DRIVE was evaluated on a benchmark dataset, which showed a better overall performance compared to other state-of-the-art methodologies, however, considerable care must be taken due to the reduced size of the dataset. DRIVE outlines the outstanding potential that multiple levels of a feature-based learning model will play in the future of oncology-based precision medicine.

6.
Nat Genet ; 52(3): 306-319, 2020 03.
Article in English | MEDLINE | ID: mdl-32024998

ABSTRACT

About half of all cancers have somatic integrations of retrotransposons. Here, to characterize their role in oncogenesis, we analyzed the patterns and mechanisms of somatic retrotransposition in 2,954 cancer genomes from 38 histological cancer subtypes within the framework of the Pan-Cancer Analysis of Whole Genomes (PCAWG) project. We identified 19,166 somatically acquired retrotransposition events, which affected 35% of samples and spanned a range of event types. Long interspersed nuclear element (LINE-1; L1 hereafter) insertions emerged as the first most frequent type of somatic structural variation in esophageal adenocarcinoma, and the second most frequent in head-and-neck and colorectal cancers. Aberrant L1 integrations can delete megabase-scale regions of a chromosome, which sometimes leads to the removal of tumor-suppressor genes, and can induce complex translocations and large-scale duplications. Somatic retrotranspositions can also initiate breakage-fusion-bridge cycles, leading to high-level amplification of oncogenes. These observations illuminate a relevant role of L1 retrotransposition in remodeling the cancer genome, with potential implications for the development of human tumors.


Subject(s)
Carcinogenesis/genetics , Gene Rearrangement/genetics , Genome, Human/genetics , Long Interspersed Nucleotide Elements/genetics , Neoplasms/genetics , Retroelements/genetics , Humans , Neoplasms/pathology
7.
Nat Genet ; 51(3): 506-516, 2019 03.
Article in English | MEDLINE | ID: mdl-30718927

ABSTRACT

Esophageal adenocarcinoma (EAC) is a poor-prognosis cancer type with rapidly rising incidence. Understanding of the genetic events driving EAC development is limited, and there are few molecular biomarkers for prognostication or therapeutics. Using a cohort of 551 genomically characterized EACs with matched RNA sequencing data, we discovered 77 EAC driver genes and 21 noncoding driver elements. We identified a mean of 4.4 driver events per tumor, which were derived more commonly from mutations than copy number alterations, and compared the prevelence of these mutations to the exome-wide mutational excess calculated using non-synonymous to synonymous mutation ratios (dN/dS). We observed mutual exclusivity or co-occurrence of events within and between several dysregulated EAC pathways, a result suggestive of strong functional relationships. Indicators of poor prognosis (SMAD4 and GATA4) were verified in independent cohorts with significant predictive value. Over 50% of EACs contained sensitizing events for CDK4 and CDK6 inhibitors, which were highly correlated with clinically relevant sensitivity in a panel of EAC cell lines and organoids.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Esophageal Neoplasms/genetics , Cohort Studies , DNA Copy Number Variations/genetics , Exome/genetics , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Genomics/methods , Humans , Male , Mutation/genetics
8.
Gastroenterology ; 153(3): 657-673.e1, 2017 09.
Article in English | MEDLINE | ID: mdl-28716721

ABSTRACT

We have recently gained unprecedented insight into genetic factors that determine risk for Barrett's esophagus (BE) and progression to esophageal adenocarcinoma (EA). Next-generation sequencing technologies have allowed us to identify somatic mutations that initiate BE and track genetic changes during development of tumors and invasive cancer. These technologies led to identification of mechanisms of tumorigenesis that challenge the current multistep model of progression to EA. Newer, cost-effective technologies create opportunities to rapidly translate the analysis of DNA into tools that can identify patients with BE at high risk for cancer, detect dysplastic lesions more reliably, and uncover mechanisms of carcinogenesis.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Carcinogenesis/genetics , Esophageal Neoplasms/genetics , Mutation , Adenocarcinoma/diagnosis , Adenocarcinoma/drug therapy , DNA Copy Number Variations , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/drug therapy , Genome-Wide Association Study , Germ-Line Mutation , Humans
10.
F1000Res ; 5: 1336, 2016.
Article in English | MEDLINE | ID: mdl-27594985

ABSTRACT

Esophageal adenocarcinoma (EAC) is highly mutated and molecularly heterogeneous. The number of cell lines available for study is limited and their genome has been only partially characterized. The availability of an accurate annotation of their mutational landscape is crucial for accurate experimental design and correct interpretation of genotype-phenotype findings. We performed high coverage, paired end whole genome sequencing on eight EAC cell lines-ESO26, ESO51, FLO-1, JH-EsoAd1, OACM5.1 C, OACP4 C, OE33, SK-GT-4-all verified against original patient material, and one esophageal high grade dysplasia cell line, CP-D. We have made available the aligned sequence data and report single nucleotide variants (SNVs), small insertions and deletions (indels), and copy number alterations, identified by comparison with the human reference genome and known single nucleotide polymorphisms (SNPs). We compare these putative mutations to mutations found in primary tissue EAC samples, to inform the use of these cell lines as a model of EAC.

11.
Nat Genet ; 48(10): 1131-41, 2016 10.
Article in English | MEDLINE | ID: mdl-27595477

ABSTRACT

Esophageal adenocarcinoma (EAC) has a poor outcome, and targeted therapy trials have thus far been disappointing owing to a lack of robust stratification methods. Whole-genome sequencing (WGS) analysis of 129 cases demonstrated that this is a heterogeneous cancer dominated by copy number alterations with frequent large-scale rearrangements. Co-amplification of receptor tyrosine kinases (RTKs) and/or downstream mitogenic activation is almost ubiquitous; thus tailored combination RTK inhibitor (RTKi) therapy might be required, as we demonstrate in vitro. However, mutational signatures showed three distinct molecular subtypes with potential therapeutic relevance, which we verified in an independent cohort (n = 87): (i) enrichment for BRCA signature with prevalent defects in the homologous recombination pathway; (ii) dominant T>G mutational pattern associated with a high mutational load and neoantigen burden; and (iii) C>A/T mutational pattern with evidence of an aging imprint. These subtypes could be ascertained using a clinically applicable sequencing strategy (low coverage) as a basis for therapy selection.


Subject(s)
Adenocarcinoma/genetics , Esophageal Neoplasms/genetics , Mutation , Adenocarcinoma/classification , Adenocarcinoma/immunology , Adenocarcinoma/therapy , Aged , Antineoplastic Agents/therapeutic use , CD8 Antigens/immunology , Cell Line, Tumor , Cohort Studies , DNA Damage , DNA, Neoplasm , Esophageal Neoplasms/classification , Esophageal Neoplasms/immunology , Esophageal Neoplasms/therapy , Female , Genetic Heterogeneity , Genome, Human , Humans , Male , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Sequence Analysis, DNA
12.
New Bioeth ; 22(1): 45-55, 2016 Apr.
Article in English | MEDLINE | ID: mdl-28219279

ABSTRACT

Public, occupational and environmental health are relatively novel disciplines compared to the ancient history of medicine. Their development, together with a more insightful knowledge of the human pathophysiology (this more usual term is the one used in the article itself), have progressively expanded the field of investigation of medicine to environmental, behavioural and genetic factors that favour the development of certain medical conditions. As a result we have developed numerous additional strategies to monitor health and prevent disease, including interventions in anticipation of diseases themselves when patients are still healthy or in a grey area of increased risk. New developments related to genomics and distributed point of care technologies will exacerbate a process of medicalization of health. This process is profoundly re-shaping how medicine interacts with the general population, states and policy makers and has implications for healthcare system design and individual health choices.


Subject(s)
Delivery of Health Care , Medicalization , Humans , Medicine , Risk
13.
Clin Cancer Res ; 21(2): 396-404, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25348516

ABSTRACT

PURPOSE: Improved therapeutic approaches are needed for the treatment of pancreatic ductal adenocarcinoma (PDAC). As dual MEK and PI3K inhibition is presently being used in clinical trials for patients with PDAC, we sought to test the efficacy of combined targeting of these pathways in PDAC using both in vitro drug screens and genetically engineered mouse models (GEMM). EXPERIMENTAL DESIGN: We performed high-throughput screening of >500 human cancer cell lines (including 46 PDAC lines), for sensitivity to 50 clinically relevant compounds, including MEK and PI3K inhibitors. We tested the top hit in the screen, the MEK1/2 inhibitor, AZD6244, for efficacy alone or in combination with the PI3K inhibitors, BKM120 or GDC-0941, in a Kras(G12D)-driven GEMM that recapitulates the histopathogenesis of human PDAC. RESULTS: In vitro screens revealed that PDAC cell lines are relatively resistant to single-agent therapies. The response profile to the MEK1/2 inhibitor, AZD6244, was an outlier, showing the highest selective efficacy in PDAC. Although MEK inhibition alone was mainly cytostatic, apoptosis was induced when combined with PI3K inhibitors (BKM120 or GDC-0941). When tested in a PDAC GEMM and compared with the single agents or vehicle controls, the combination delayed tumor formation in the setting of prevention and extended survival when used to treat advanced tumors, although no durable responses were observed. CONCLUSIONS: Our studies point to important contributions of MEK and PI3K signaling to PDAC pathogenesis and suggest that dual targeting of these pathways may provide benefit in some patients with PDAC. Clin Cancer Res; 21(2); 396-404. ©2014 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Aminopyridines , Animals , Benzimidazoles/pharmacology , Cell Line, Tumor , Disease Models, Animal , Drug Screening Assays, Antitumor , Drug Synergism , Erlotinib Hydrochloride , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mice, Transgenic , Morpholines , Phosphoinositide-3 Kinase Inhibitors , Quinazolines/pharmacology
14.
J Clin Invest ; 123(2): 727-39, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23321669

ABSTRACT

Epigenetic mechanisms mediate heritable control of cell identity in normal cells and cancer. We sought to identify epigenetic regulators driving the pathogenesis of pancreatic ductal adenocarcinoma (PDAC), one of the most lethal human cancers. We found that KDM2B (also known as Ndy1, FBXL10, and JHDM1B), an H3K36 histone demethylase implicated in bypass of cellular senescence and somatic cell reprogramming, is markedly overexpressed in human PDAC, with levels increasing with disease grade and stage, and highest expression in metastases. KDM2B silencing abrogated tumorigenicity of PDAC cell lines exhibiting loss of epithelial differentiation, whereas KDM2B overexpression cooperated with KrasG12D to promote PDAC formation in mouse models. Gain- and loss-of-function experiments coupled to genome-wide gene expression and ChIP studies revealed that KDM2B drives tumorigenicity through 2 different transcriptional mechanisms. KDM2B repressed developmental genes through cobinding with Polycomb group (PcG) proteins at transcriptional start sites, whereas it activated a module of metabolic genes, including mediators of protein synthesis and mitochondrial function, cobound by the MYC oncogene and the histone demethylase KDM5A. These results defined epigenetic programs through which KDM2B subverts cellular differentiation and drives the pathogenesis of an aggressive subset of PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/etiology , F-Box Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Oxidoreductases, N-Demethylating/metabolism , Pancreatic Neoplasms/etiology , Polycomb-Group Proteins/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Disease Models, Animal , Epigenesis, Genetic , F-Box Proteins/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Mice, SCID , Oxidoreductases, N-Demethylating/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Polycomb-Group Proteins/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Transcription, Genetic , Up-Regulation
15.
Surg Obes Relat Dis ; 9(5): 696-700, 2013.
Article in English | MEDLINE | ID: mdl-22951077

ABSTRACT

BACKGROUND: In an effort to provide better cosmesis for patients, there has been a surge recently in the use of laparoendoscopic single-site adjustable gastric banding. Few data, however, are available on the long-term wound complications resulting from this technique. We conducted a retrospective review of patients to identify the extent of wound complications found during a minimum follow-up period of 2 years after laparoendoscopic single-site adjustable gastric banding. The complications evaluated included infection, hernia rates, and port and tubing complications. All the laparoendoscopic single-site adjustable gastric banding cases were performed at University of Illinois Medical Center by a single surgeon. METHODS: Twenty-five patients underwent single-site laparoscopic adjustable gastric banding from March 2009 to January 2010, and the data were reviewed retrospectively. The single incision was made with multifascial trocar placement using conventional laparoscopic instruments. The patients were followed up during band adjustments and clinic visits and by telephone interview. RESULTS: Six months after surgery, 1 patient required port removal because of port site infection with internalization of the tubing. A second patient experienced a foul-smelling, clear discharge and was treated with antibiotics, with no additional consequences. No incisional hernias or flipped ports were noted. CONCLUSION: In our experience, laparoendoscopic single-site adjustable gastric banding produced a low rate of port and wound site complications in patients during a minimum follow-up period of 2 years. We believe this is a valid alternative to the standard procedure, providing cosmetic advantages and a low wound complication rate in morbidly obese patients.


Subject(s)
Gastroplasty/methods , Laparoscopy/methods , Obesity, Morbid/surgery , Postoperative Complications/epidemiology , Adolescent , Adult , Female , Follow-Up Studies , Hernia/epidemiology , Humans , Illinois/epidemiology , Male , Middle Aged , Retrospective Studies , Surgical Wound Infection/epidemiology
16.
World J Surg Oncol ; 10: 184, 2012 Sep 10.
Article in English | MEDLINE | ID: mdl-22963172

ABSTRACT

BACKGROUND: Surgery is still the standard treatment for aggressive fibromatosis (AF); however, local control remains a significant problem and the impact of R0 surgery on cumulative recurrence (CR) is objective of contradictory reports. METHODS: This is a single-institution study of 62 consecutive patients affected by extra-abdominal and intra-abdominal AF who received macroscopically radical surgery within a time period of 15 years. RESULTS: Definitive pathology examination confirmed an R0 situation in 49 patients and an R1 in 13 patients. Five-year CR for patients who underwent R0 vs R1 surgery was 7.1% vs 46.4% (P = 0.04) and for limbs vs other localizations 33.3% vs 9.9% (P = 0.02) respectively. In 17 patients who had intraoperative frozen section (IFS) margin evaluation R0 surgery was more common (17 of 17 vs 32 of 45, P = 0.01) and CR lower (five-year CR 0% vs 19.1%, respectively, P = 0.04). However, in multivariate analysis only limb localization showed a negative impact on CR (HR: 1.708, 95% CI 1.03 to 2.84, P = 0.04). CONCLUSIONS: IFS evaluation could help the surgeon to achieve R0 surgery in AF. Non-surgical treatment, including watchful follow-up, could be indicated for patients with limb AF localization, because of their high risk of recurrence even after R0 surgery.


Subject(s)
Fibromatosis, Aggressive/surgery , Neoplasm Recurrence, Local/etiology , Neoplasm, Residual/etiology , Adolescent , Adult , Aged , Female , Fibromatosis, Aggressive/mortality , Fibromatosis, Aggressive/pathology , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/mortality , Neoplasm Staging , Neoplasm, Residual/diagnosis , Neoplasm, Residual/mortality , Prognosis , Remission Induction , Risk Factors , Survival Rate , Young Adult
17.
Nature ; 487(7408): 510-3, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22763454

ABSTRACT

Circulating tumour cells (CTCs) shed into blood from primary cancers include putative precursors that initiate distal metastases. Although these cells are extraordinarily rare, they may identify cellular pathways contributing to the blood-borne dissemination of cancer. Here, we adapted a microfluidic device for efficient capture of CTCs from an endogenous mouse pancreatic cancer model and subjected CTCs to single-molecule RNA sequencing, identifying Wnt2 as a candidate gene enriched in CTCs. Expression of WNT2 in pancreatic cancer cells suppresses anoikis, enhances anchorage-independent sphere formation, and increases metastatic propensity in vivo. This effect is correlated with fibronectin upregulation and suppressed by inhibition of MAP3K7 (also known as TAK1) kinase. In humans, formation of non-adherent tumour spheres by pancreatic cancer cells is associated with upregulation of multiple WNT genes, and pancreatic CTCs revealed enrichment for WNT signalling in 5 out of 11 cases. Thus, molecular analysis of CTCs may identify candidate therapeutic targets to prevent the distal spread of cancer.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Neoplasm Metastasis/genetics , Neoplastic Cells, Circulating/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Wnt Proteins/metabolism , Wnt Signaling Pathway/genetics , Animals , Cell Survival , Contact Inhibition , Disease Models, Animal , Genes, Neoplasm/genetics , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mice , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Sequence Analysis, RNA , Wnt Proteins/genetics , Wnt2 Protein/genetics , Wnt2 Protein/metabolism
18.
Cancer Res ; 72(18): 4840-5, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22787119

ABSTRACT

The TGF-ß pathway is under active consideration as a cancer drug target based on its capacity to promote cancer cell invasion and to create a protumorigenic microenvironment. However, the clinical application of TGF-ß inhibitors remains uncertain as genetic studies show a tumor suppressor function of TGF-ß in pancreatic cancer and other epithelial malignancies. Here, we used genetically engineered mouse models to investigate the therapeutic impact of global TGF-ß inhibition in pancreatic cancer in relation to tumor stage, genetic profile, and concurrent chemotherapy. We found that αvß6 integrin acted as a key upstream activator of TGF-ß in evolving pancreatic cancers. In addition, TGF-ß or αvß6 blockade increased tumor cell proliferation and accelerated both early and later disease stages. These effects were dependent on the presence of Smad4, a central mediator of TGF-ß signaling. Therefore, our findings indicate that αvß6 and TGF-ß act in a common tumor suppressor pathway whose pharmacologic inactivation promotes pancreatic cancer progression.


Subject(s)
Antigens, Neoplasm/metabolism , Integrins/metabolism , Pancreatic Neoplasms/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Animals , Disease Models, Animal , Disease Progression , Immunohistochemistry , Mice
19.
J Biol Chem ; 286(38): 33061-9, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21757686

ABSTRACT

Sustained expression of the histone demethylase, KDM2B (Ndy1/FBXL10/JHDM1B), bypasses cellular senescence in primary mouse embryonic fibroblasts (MEFs). Here, we show that KDM2B is a conserved regulator of lifespan in multiple primary cell types and defines a program in which this chromatin-modifying enzyme counteracts the senescence-associated down-regulation of the EZH2 histone methyltransferase. Senescence in MEFs epigenetically silences KDM2B and induces the tumor suppressor miRNAs let-7b and miR-101, which target EZH2. Forced expression of KDM2B promotes immortalization by silencing these miRNAs through locus-specific histone H3 K36me2 demethylation, leading to EZH2 up-regulation. Overexpression of let-7b down-regulates EZH2, induces premature senescence, and counteracts immortalization of MEFs driven by KDM2B. The KDM2B-let-7-EZH2 pathway also contributes to the proliferation of immortal Ink4a/Arf null fibroblasts suggesting that, beyond its anti-senescence role in primary cells, this histone-modifying enzyme functions more broadly in the regulation of cellular proliferation.


Subject(s)
Cell Cycle , Cellular Senescence , F-Box Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine/metabolism , MicroRNAs/metabolism , Signal Transduction , ADP-Ribosylation Factors/metabolism , Animals , Cell Proliferation , Cells, Cultured , Conserved Sequence/genetics , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mice, Inbred C57BL , Polycomb Repressive Complex 2 , Substrate Specificity , Up-Regulation/genetics
20.
Cancer Res ; 71(14): 5020-9, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21586612

ABSTRACT

The STAT3 transcription factor is an important regulator of stem cell self-renewal, cancer cell survival, and inflammation. In the pancreas, STAT3 is dispensable for normal development, whereas the majority of pancreatic ductal adenocarcinomas (PDAC) show constitutive activation of STAT3, suggesting its potential as a therapeutic target in this cancer. Here, we sought to define the mechanisms of STAT3 activation and its functional importance in PDAC pathogenesis. Large-scale screening of cancer cell lines with a JAK2 inhibitor that blocks STAT3 function revealed a more than 30-fold range in sensitivity in PDAC, and showed a close correlation of sensitivity with levels of tyrosine-phosphorylated STAT3 and of the gp130 receptor, an upstream signaling component. Correspondingly, upregulation of the IL6/LIF-gp130 pathway accounted for the strong STAT3 activation in PDAC subsets. To define functions of STAT3 in vivo, we developed mouse models that test the impact of conditional inactivation of STAT3 in KRAS-driven PDAC. We showed that STAT3 is required for the development of the earliest premalignant pancreatic lesions, acinar-to-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia (PanIN). Moreover, acute STAT3 inactivation blocked PDAC initiation in a second in vivo model. Our results show that STAT3 has critical roles throughout the course of PDAC pathogenesis, supporting the development of therapeutic approaches targeting this pathway. Moreover, our work suggests that gp130 and phospho-STAT3 expression may be effective biomarkers for predicting response to JAK2 inhibitors.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Cell Transformation, Neoplastic/genetics , Genes, ras , Pancreatic Neoplasms/genetics , Precancerous Conditions/genetics , STAT3 Transcription Factor/genetics , Animals , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Genetic Predisposition to Disease , Humans , Mice , Mice, SCID , Mice, Transgenic , Pancreatic Neoplasms/metabolism , Phosphorylation , Precancerous Conditions/metabolism , STAT3 Transcription Factor/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...