Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
2.
J Clin Anesth ; 89: 111196, 2023 10.
Article in English | MEDLINE | ID: mdl-37406462

ABSTRACT

STUDY OBJECTIVE: To determine if a nasal positive airway pressure (nasal CPAP) mask would decrease the number of hypoxemic events in obese and obstructive sleep apnea patients undergoing colonoscopy. DESIGN: Single-center prospective randomized controlled trial. SETTING: Tertiary academic center. PATIENTS: We enrolled 109 patients with diagnosis of obesity and/or obstructive sleep apnea scheduled to undergo colonoscopy under propofol general anesthesia without planned tracheal intubation. INTERVENTION: Patients were randomly allocated (1:1 ratio) to receive supplementary oxygen at a flow of 10 L/min, either through a nasal CPAP or a simple facemask. MEASUREMENTS: The primary endpoint was the difference in the mean percentage of time spent with oxygen saturation below 90% between the two groups. Secondary outcomes included the need for airway maneuvers/interventions, average SpO2 during the case, duration and severity of oxygen desaturation, incidence and duration of procedural interruptions, and satisfaction and tolerance scores. MAIN RESULTS: 54 were allocated to the simple face mask and 55 to the nasal CPAP mask arms, respectively. A total of 6 patients experienced a hypoxemic event. Among these patients, the difference in the percentage of time spent with oxygen saturation below 90% was not clinically relevant (p = 1.0). However, patients in the nasal CPAP group required less chin lift (20% vs. 42.6%; p = 0.01) and oral cannula insertion (12.7% vs.29.6%; p = 0.03). The percentage of patients with at least one airway maneuver was higher in the simple face mask arm (68.5% vs. 41.8%; p = 0.005). Patient tolerance to device score was lower in the nasal CPAP group (8.85 vs. 9.56; p = 0.003). CONCLUSIONS: A nasal CPAP did not prevent hypoxemia and should not be used routinely for colonoscopy in obese or OSA patients if a simple face mask is an alternative therapy. However, potential advantages of its use include fewer airway maneuvers or interventions, which may be desirable in certain clinical settings. TRIAL REGISTRATION: Clinicaltrials.gov, identifier: NCT05175573.


Subject(s)
Propofol , Sleep Apnea, Obstructive , Humans , Adult , Masks/adverse effects , Prospective Studies , Sleep Apnea, Obstructive/therapy , Sleep Apnea, Obstructive/etiology , Obesity/complications , Oxygen , Intubation, Intratracheal , Anesthesia, General , Colonoscopy , Continuous Positive Airway Pressure
3.
Development ; 150(2)2023 01 15.
Article in English | MEDLINE | ID: mdl-36458554

ABSTRACT

Adenosine deaminase acting on RNA 1 (ADAR1) is an RNA-binding protein that deaminates adenosine (A) to inosine (I). A-to-I editing alters post-transcriptional RNA processing, making ADAR1 a crucial regulator of gene expression. Consequently, Adar1 has been implicated in organogenesis. To determine the role of Adar1 in pancreatic development and homeostasis, we conditionally deleted Adar1 from the murine pancreas (Ptf1aCre/+; Adar1Fl/Fl). The resulting mice had stunted growth, likely due to malabsorption associated with exocrine pancreatic insufficiency. Analyses of pancreata revealed ductal cell expansion, heightened interferon-stimulated gene expression and an increased influx of immune cells. Concurrent deletion of Adar1 and Mavs, a signaling protein implicated in the innate immune pathway, rescued the degenerative phenotype and resulted in normal pancreatic development. Taken together, our work suggests that the primary function of Adar1 in the pancreas is to prevent aberrant activation of the Mavs-mediated innate immune pathway, thereby maintaining pancreatic homeostasis.


Subject(s)
Pancreas, Exocrine , Animals , Mice , Pancreas, Exocrine/metabolism , Interferons/genetics , Interferons/metabolism , Phenotype , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism
5.
Cancer Res ; 82(15): 2761-2776, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35666804

ABSTRACT

Conventional genetically engineered mouse models (GEMM) are time-consuming, laborious, and offer limited spatiotemporal control. Here, we describe the development of a streamlined platform for in vivo gene activation using CRISPR activation (CRISPRa) technology. Unlike conventional GEMMs, this model system allows for flexible, sustained, and timed activation of one or more target genes using single or pooled lentiviral guides. Myc and Yap1 were used as model oncogenes to demonstrate gene activation in primary pancreatic organoid cultures in vitro and enhanced tumorigenic potential in Myc-activated organoids when transplanted orthotopically in vivo. Implementation of this model as an autochthonous lung cancer model showed that transduction-mediated activation of Myc led to accelerated tumor progression and significantly reduced overall survival relative to nontargeted tumor controls. Furthermore, Myc activation led to the acquisition of an immune suppressive, "cold" tumor microenvironment. Cross-species validation of these results using publicly available RNA/DNA-seq datasets linked MYC to a previously described immunosuppressive molecular subtype in patient tumors, thus identifying a patient cohort that may benefit from combined MYC- and immune-targeted therapies. Overall, this work demonstrates how CRISPRa can be used for rapid functional validation of putative oncogenes and may allow for the identification and evaluation of potential metastatic and oncogenic drivers through competitive screening. SIGNIFICANCE: A streamlined platform for programmable CRISPR gene activation enables rapid evaluation and functional validation of putative oncogenes in vivo.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Proto-Oncogene Proteins c-myc , Adenocarcinoma of Lung/genetics , Animals , Carcinogenesis/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Oncogenes/genetics , Proto-Oncogene Proteins c-myc/genetics , Tumor Microenvironment/genetics
6.
Eur J Gastroenterol Hepatol ; 34(1): 33-38, 2022 01 01.
Article in English | MEDLINE | ID: mdl-33470698

ABSTRACT

BACKGROUND: Early detection of pancreatic ductal adenocarcinoma (PDA) may improve survival. We previously developed a clinical prediction model among patients with new-onset diabetes to help identify PDAs 6 months prior to the clinical diagnosis of the cancer. We developed and internally validated a new model to predict PDA risk among those newly diagnosed with impaired fasting glucose (IFG). METHODS: We conducted a retrospective cohort study in The Health Improvement Network (THIN) (1995-2013) from the UK. Eligible study patients had newly diagnosed IFG during follow-up in THIN. The outcome was incident PDA diagnosed within 3 years of IFG diagnosis. Candidate predictors were factors associated with PDA, glucose metabolism or both. RESULTS: Among the 138 232 eligible patients with initial IFG diagnosis, 245 (0.2%) were diagnosed with PDA within 3 years. The median time from IFG diagnosis to clinical PDA diagnosis was 326 days (IQR 120-588). The final prediction model included age, BMI, proton pump inhibitor use, total cholesterol, low-density lipoprotein, alanine aminotransferase and alkaline phosphatase. The model achieved good discrimination [area under the curve 0.71 (95% CI, 0.67-0.75)] and calibration (Hosmer and Lemeshow goodness-of-fit test P > 0.05 in 17 of the 20 imputed data sets) with optimism of 0.0012662 (95% CI, -0.00932 to 0.0108771). CONCLUSIONS: We developed and internally validated a sequential PDA prediction model based on clinical information routinely available at the initial appearance of IFG. If externally validated, this model could significantly extend our ability to detect PDAs at an earlier stage.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Prediabetic State , Blood Glucose , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/epidemiology , Humans , Models, Statistical , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/epidemiology , Prediabetic State/diagnosis , Prediabetic State/epidemiology , Prognosis , Retrospective Studies , Pancreatic Neoplasms
7.
Clin Transl Gastroenterol ; 12(11): e00431, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34797250

ABSTRACT

INTRODUCTION: Chronic pancreatitis is associated with an increased risk of developing pancreatic cancer, and patients with inherited forms of pancreatitis are at greatest risk. We investigated whether clinical severity of pancreatitis could also be an indicator of cancer risk independent of etiology by performing targeted DNA sequencing to assess the mutational burden in 55 cancer-associated genes. METHODS: Using picodroplet digital polymerase chain reaction and next-generation sequencing, we reported the genomic profiles of pancreases from severe clinical cases of chronic pancreatitis that necessitated palliative total pancreatectomy with islet autotransplantation. RESULTS: We assessed 57 tissue samples from 39 patients with genetic and idiopathic etiologies and found that despite the clinical severity of disease, there was no corresponding increase in mutational burden. The average allele frequency of somatic variants was 1.19% (range 1.00%-5.97%), and distinct regions from the same patient displayed genomic heterogeneity, suggesting that these variants are subclonal. Few oncogenic KRAS mutations were discovered (7% of all samples), although we detected evidence of frequent cancer-related variants in other genes such as TP53, CDKN2A, and SMAD4. Of note, tissue samples with oncogenic KRAS mutations and samples from patients with PRSS1 mutations harbored an increased total number of somatic variants, suggesting that these patients may have increased genomic instability and could be at an increased risk of developing pancreatic cancer. DISCUSSION: Overall, we showed that even in those patients with chronic pancreatitis severe enough to warrant total pancreatectomy with islet autotransplantation, pancreatic cancer-related mutational burden is not appreciably increased.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Mutation , Pancreatic Neoplasms/genetics , Pancreatitis, Chronic/genetics , Adult , Age of Onset , Child , Female , Humans , Islets of Langerhans Transplantation , Male , Pancreatectomy , Pancreatitis, Chronic/complications , Pancreatitis, Chronic/surgery , Patient Acuity , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins p21(ras)/genetics , Trypsin/genetics
8.
Science ; 373(6561): eabj0486, 2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34529467

ABSTRACT

Inflammation is a major risk factor for pancreatic ductal adenocarcinoma (PDAC). When occurring in the context of pancreatitis, KRAS mutations accelerate tumor development in mouse models. We report that long after its complete resolution, a transient inflammatory event primes pancreatic epithelial cells to subsequent transformation by oncogenic KRAS. Upon recovery from acute inflammation, pancreatic epithelial cells display an enduring adaptive response associated with sustained transcriptional and epigenetic reprogramming. Such adaptation enables the reactivation of acinar-to-ductal metaplasia (ADM) upon subsequent inflammatory events, thereby limiting tissue damage through a rapid decrease of zymogen production. We propose that because activating mutations of KRAS maintain an irreversible ADM, they may be beneficial and under strong positive selection in the context of recurrent pancreatitis.


Subject(s)
Acinar Cells/pathology , Carcinogenesis , Carcinoma, Pancreatic Ductal/pathology , Genes, ras , Pancreas/pathology , Pancreatitis/physiopathology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/physiopathology , Cell Transformation, Neoplastic , Cells, Cultured , Cellular Reprogramming , Chromatin/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Enzyme Precursors/metabolism , Epigenesis, Genetic , Epithelial Cells/pathology , Epithelial Cells/physiology , Female , MAP Kinase Signaling System , Male , Metaplasia , Mice , Mutation , Pancreas/metabolism , Pancreatitis/genetics , Pancreatitis/immunology , Spheroids, Cellular , Transcriptome
9.
Cell Rep ; 35(2): 108990, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852841

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is therapeutically recalcitrant and metastatic. Partial epithelial to mesenchymal transition (EMT) is associated with metastasis; however, a causal connection needs further unraveling. Here, we use single-cell RNA sequencing and genetic mouse models to identify the functional roles of partial EMT and epithelial stabilization in PDAC growth and metastasis. A global EMT expression signature identifies ∼50 cancer cell clusters spanning the epithelial-mesenchymal continuum in both human and murine PDACs. The combined genetic suppression of Snail and Twist results in PDAC epithelial stabilization and increased liver metastasis. Genetic deletion of Zeb1 in PDAC cells also leads to liver metastasis associated with cancer cell epithelial stabilization. We demonstrate that epithelial stabilization leads to the enhanced collective migration of cancer cells and modulation of the immune microenvironment, which likely contribute to efficient liver colonization. Our study provides insights into the diverse mechanisms of metastasis in pancreatic cancer and potential therapeutic targets.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Epithelial-Mesenchymal Transition/genetics , Liver Neoplasms/genetics , Nuclear Proteins/genetics , Pancreatic Neoplasms/genetics , Snail Family Transcription Factors/genetics , Twist-Related Protein 1/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/secondary , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Male , Mice , Nuclear Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Single-Cell Analysis , Snail Family Transcription Factors/metabolism , Survival Analysis , Tumor Microenvironment/genetics , Twist-Related Protein 1/metabolism , Zinc Finger E-box-Binding Homeobox 1/deficiency
10.
Nat Cancer ; 2(12): 1338-1356, 2021 12.
Article in English | MEDLINE | ID: mdl-35121902

ABSTRACT

Despite efforts in understanding its underlying mechanisms, the etiology of chromosomal instability (CIN) remains unclear for many tumor types. Here, we identify CIN initiation as a previously undescribed function for APOBEC3A (A3A), a cytidine deaminase upregulated across cancer types. Using genetic mouse models of pancreatic ductal adenocarcinoma (PDA) and genomics analyses in human tumor cells we show that A3A-induced CIN leads to aggressive tumors characterized by enhanced early dissemination and metastasis in a STING-dependent manner and independently of the canonical deaminase functions of A3A. We show that A3A upregulation recapitulates numerous copy number alterations commonly observed in patients with PDA, including co-deletions in DNA repair pathway genes, which in turn render these tumors susceptible to poly (ADP-ribose) polymerase inhibition. Overall, our results demonstrate that A3A plays an unexpected role in PDA as a specific driver of CIN, with significant effects on disease progression and treatment.


Subject(s)
Cytidine Deaminase , Pancreatic Neoplasms , Animals , Chromosomal Instability/genetics , Cytidine Deaminase/genetics , Humans , Mice , Pancreatic Neoplasms/genetics , Proteins/genetics , Pancreatic Neoplasms
11.
Gastroenterology ; 158(5): 1417-1432.e11, 2020 04.
Article in English | MEDLINE | ID: mdl-31843590

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy that invades surrounding structures and metastasizes rapidly. Although inflammation is associated with tumor formation and progression, little is known about the mechanisms of this connection. We investigate the effects of interleukin (IL) 22 in the development of pancreatic tumors in mice. METHODS: We performed studies with Pdx1-Cre;LSL-KrasG12D;Trp53+/-;Rosa26EYFP/+ (PKCY) mice, which develop pancreatic tumors, and PKCY mice with disruption of IL22 (PKCY Il22-/-mice). Pancreata were collected at different stages of tumor development and analyzed by immunohistochemistry, immunoblotting, real-time polymerase chain reaction, and flow cytometry. Some mice were given cerulean to induce pancreatitis. Pancreatic cancer cell lines (PD2560) were orthotopically injected into C57BL/6 mice or Il22-/-mice, and tumor development was monitored. Pancreatic cells were injected into the tail veins of mice, and lung metastases were quantified. Acini were collected from C57BL/6 mice and resected human pancreata and were cultured. Cell lines and acini cultures were incubated with IL22 and pharmacologic inhibitors, and protein levels were knocked down with small hairpin RNAs. We performed immunohistochemical analyses of 26 PDACs and 5 nonneoplastic pancreas specimens. RESULTS: We observed increased expression of IL22 and the IL22 receptor (IL22R) in the pancreas compared with other tissues in mice; IL22 increased with pancreatitis and tumorigenesis. Flow cytometry indicated that the IL22 was produced primarily by T-helper 22 cells. PKCY Il22-/-mice did not develop precancerous lesions or pancreatic tumors. The addition of IL22 to cultured acinar cells increased their expression of markers of ductal metaplasia; these effects of IL22 were prevented with inhibitors of Janus kinase signaling to signal transducer and activator of transcription (STAT) (ruxolitinib) or mitogen-activated protein kinase kinase (MEK) (trametinib) and with STAT3 knockdown. Pancreatic cells injected into Il22-/- mice formed smaller tumors than those injected into C57BL/6. Incubation of IL22R-expressing PDAC cells with IL22 promoted spheroid formation and invasive activity, resulting in increased expression of stem-associated transcription factors (GATA4, SOX2, SOX17, and NANOG), and increased markers of the epithelial-mesenchymal transition (CDH1, SNAI2, TWIST1, and beta catenin); ruxolitinib blocked these effects. Human PDAC tissues had higher levels of IL22, phosphorylated STAT3, and markers of the epithelial-mesenchymal transition than nonneoplastic tissues. An increased level of STAT3 in IL22R-positive cells was associated with shorter survival times of patients. CONCLUSIONS: We found levels of IL22 to be increased during pancreatitis and pancreatic tumor development and to be required for tumor development and progression in mice. IL22 promotes acinar to ductal metaplasia, stem cell features, and increased expression of markers of the epithelial-mesenchymal transition; inhibitors of STAT3 block these effects. Increased expression of IL22 by PDACs is associated with reduced survival times.


Subject(s)
Acinar Cells/pathology , Carcinoma, Pancreatic Ductal/immunology , Cell Transformation, Neoplastic/immunology , Interleukins/metabolism , Pancreatic Neoplasms/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction/immunology , Acinar Cells/immunology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor/transplantation , Cell Plasticity/drug effects , Cell Plasticity/immunology , Cell Transformation, Neoplastic/drug effects , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/immunology , Female , HEK293 Cells , Humans , Interleukins/immunology , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Male , Metaplasia/immunology , Metaplasia/pathology , Mice , Mice, Knockout , Nitriles , Pancreas/cytology , Pancreas/immunology , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pancreatitis/immunology , Pancreatitis/pathology , Pyrazoles/pharmacology , Pyridones/pharmacology , Pyrimidines , Pyrimidinones/pharmacology , RNA, Small Interfering/metabolism , Receptors, Interleukin/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Survival Analysis , Interleukin-22
12.
Genes Dev ; 33(11-12): 641-655, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31048544

ABSTRACT

Pancreatic adenocarcinoma (PDA) is an aggressive disease driven by oncogenic KRAS and characterized by late diagnosis and therapeutic resistance. Here we show that deletion of the ataxia-telangiectasia group D-complementing (Atdc) gene, whose human homolog is up-regulated in the majority of pancreatic adenocarcinoma, completely prevents PDA development in the context of oncogenic KRAS. ATDC is required for KRAS-driven acinar-ductal metaplasia (ADM) and its progression to pancreatic intraepithelial neoplasia (PanIN). As a result, mice lacking ATDC are protected from developing PDA. Mechanistically, we show ATDC promotes ADM progression to PanIN through activation of ß-catenin signaling and subsequent SOX9 up-regulation. These results provide new insight into PDA initiation and reveal ATDC as a potential target for preventing early tumor-initiating events.


Subject(s)
Carcinogenesis , Carcinoma, Pancreatic Ductal/physiopathology , Pancreatic Neoplasms/physiopathology , Proto-Oncogene Proteins p21(ras)/metabolism , Transcription Factors/physiology , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Carcinoma in Situ/pathology , Carcinoma in Situ/physiopathology , Carcinoma, Pancreatic Ductal/pathology , Cell Transdifferentiation , Cells, Cultured , DNA-Binding Proteins/metabolism , Down-Regulation , Gene Knockdown Techniques , Humans , Metaplasia , Mice , Mice, Transgenic , Pancreatic Ducts/metabolism , Pancreatic Ducts/pathology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , beta Catenin/metabolism
13.
PLoS One ; 13(10): e0204875, 2018.
Article in English | MEDLINE | ID: mdl-30332430

ABSTRACT

BACKGROUND: Physical activity is associated with a lower risk of disease recurrence among colon cancer patients. Circulating tumor cells (CTC) are prognostic of disease recurrence among stage I-III colon cancer patients. The pathways through which physical activity may alter disease outcomes are unknown, but may be mediated by changes in CTCs. METHODS: Participants included 23 stage I-III colon cancer patients randomized into one of three groups: usual-care control, 150 min∙wk-1 of aerobic exercise (low-dose), and 300 min∙wk-1 of aerobic exercise (high-dose) for six months. CTCs from venous blood were quantified in a blinded fashion using an established microfluidic antibody-mediated capture device. Poisson regression models estimated the logarithmic counts of CTCs. RESULTS: At baseline, 78% (18/23) of patients had ≥1 CTC. At baseline, older age (-0.12±0.06; P = 0.04), lymphovascular invasion (0.63±0.25; P = 0.012), moderate/poor histology (1.09±0.34; P = 0.001), body mass index (0.07±0.02; P = 0.001), visceral adipose tissue (0.08±0.04; P = 0.036), insulin (0.06±0.02; P = 0.011), sICAM-1 (0.04±0.02; P = 0.037), and sVCAM-1 (0.06±0.03; P = 0.045) were associated with CTCs. Over six months, significant decreases in CTCs were observed in the low-dose (-1.34±0.34; P<0.001) and high-dose (-1.18±0.40; P = 0.004) exercise groups, whereas no significant change was observed in the control group (-0.59±0.56; P = 0.292). Over six months, reductions in body mass index (-0.07±0.02; P = 0.007), insulin (-0.08±0.03; P = 0.014), and sICAM-1 (-0.07±0.03; P = 0.005) were associated with reductions in CTCs. The main limitations of this proof-of-concept study are the small sample size, heterogenous population, and per-protocol statistical analysis. CONCLUSION: Exercise may reduce CTCs among stage I-III colon cancer patients. Changes in host factors correlated with changes in CTCs. Exercise may have a direct effect on CTCs and indirect effects through alterations in host factors. This hypothesis-generating observation derived from a small pilot study warrants further investigation and replication.


Subject(s)
Colonic Neoplasms/rehabilitation , Exercise Therapy/methods , Neoplastic Cells, Circulating/pathology , Aged , Cell Count , Colonic Neoplasms/pathology , Colonic Neoplasms/surgery , Female , Humans , Male , Middle Aged , Neoplasm Staging , Patient Compliance , Pilot Projects , Poisson Distribution , Random Allocation
14.
Dev Cell ; 45(6): 696-711.e8, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29920275

ABSTRACT

The regulation of metastatic organotropism in pancreatic ductal a denocarcinoma (PDAC) remains poorly understood. We demonstrate, using multiple mouse models, that liver and lung metastatic organotropism is dependent upon p120catenin (p120ctn)-mediated epithelial identity. Mono-allelic p120ctn loss accelerates KrasG12D-driven pancreatic cancer formation and liver metastasis. Importantly, one p120ctn allele is sufficient for E-CADHERIN-mediated cell adhesion. By contrast, cells with bi-allelic p120ctn loss demonstrate marked lung organotropism; however, rescue with p120ctn isoform 1A restores liver metastasis. In a p120ctn-independent PDAC model, mosaic loss of E-CADHERIN expression reveals selective pressure for E-CADHERIN-positive liver metastasis and E-CADHERIN-negative lung metastasis. Furthermore, human PDAC and liver metastases support the premise that liver metastases exhibit predominantly epithelial characteristics. RNA-seq demonstrates differential induction of pathways associated with metastasis and epithelial-to-mesenchymal transition in p120ctn-deficient versus p120ctn-wild-type cells. Taken together, P120CTN and E-CADHERIN mediated epithelial plasticity is an addition to the conceptual framework underlying metastatic organotropism in pancreatic cancer.


Subject(s)
Catenins/metabolism , Cell Plasticity/physiology , Pancreatic Neoplasms/pathology , Animals , Cadherins/metabolism , Cell Adhesion , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver Neoplasms/genetics , Lung Neoplasms/genetics , Mice , Neoplasm Metastasis/physiopathology , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/metabolism , Phosphoproteins/metabolism , Protein Isoforms/metabolism , Delta Catenin
15.
Psychooncology ; 27(4): 1221-1228, 2018 04.
Article in English | MEDLINE | ID: mdl-29388275

ABSTRACT

OBJECTIVE: To examine the dose-response effects of aerobic exercise on health-related quality of life (HRQoL) among colon cancer survivors. METHODS: Thirty-nine stage I to III colon cancer survivors were randomized to 1 of 3 groups: usual-care control, 150 min·wk-1 of aerobic exercise (low-dose) and 300 min·wk-1 of aerobic exercise (high-dose) for 6 months. HRQoL outcomes included the Short Form (SF)-36 physical and mental component summary, Functional Assessment of Cancer Therapy-Colorectal, Pittsburgh Sleep Quality Index, Fear of Cancer Recurrence Inventory, Fatigue Symptom Inventory, and North Central Cancer Treatment Group bowel function questionnaire, assessed at baseline and post intervention. The primary hypothesis was that exercise would improve HRQoL outcomes in a dose-response fashion, such that high-dose aerobic exercise would yield the largest improvements in HRQoL outcomes. RESULTS: Over 6 months, the low-dose group completed 141 ± 10 min·wk-1 of aerobic exercise, and the high-dose group completed 247 ± 11 min·wk-1 of aerobic exercise. Over 6 months, exercise improved the physical component summary score of the SF-36 (Ptrend  = 0.002), the Functional Assessment of Cancer Therapy-Colorectal (Ptrend  = 0.025), the Pittsburgh Sleep Quality Index (Ptrend  = 0.049), and the Fatigue Symptom Inventory (Ptrend  = 0.045) in a dose-response fashion. Between-group standardized mean difference effects sizes for the above-described findings were small to moderate in magnitude (0.35-0.75). No dose-response effects were observed for the mental component summary score of the SF-36, the Fear of Cancer Recurrence Inventory, or bowel function. CONCLUSION: Higher doses of aerobic exercise, up to 300 min·wk-1 , improve multiple HRQoL outcomes among stage I to III colon cancer survivors. These findings provide evidence that aerobic exercise may provide multiple health benefits for colon cancer survivors.


Subject(s)
Cancer Survivors/psychology , Colonic Neoplasms/psychology , Exercise Therapy/methods , Exercise/psychology , Quality of Life , Colonic Neoplasms/physiopathology , Colonic Neoplasms/rehabilitation , Exercise/physiology , Fatigue/physiopathology , Fatigue/psychology , Female , Health Status , Humans , Male , Middle Aged , Patient Reported Outcome Measures
16.
Gastroenterology ; 154(5): 1509-1523.e5, 2018 04.
Article in English | MEDLINE | ID: mdl-29273451

ABSTRACT

BACKGROUND & AIMS: Intraductal papillary mucinous neoplasias (IPMNs) are precancerous cystic lesions that can develop into pancreatic ductal adenocarcinomas (PDACs). These large macroscopic lesions are frequently detected during medical imaging, but it is unclear how they form or progress to PDAC. We aimed to identify cells that form IPMNs and mutations that promote IPMN development and progression. METHODS: We generated mice with disruption of Pten specifically in ductal cells (Sox9CreERT2;Ptenflox/flox;R26RYFP or PtenΔDuct/ΔDuct mice) and used PtenΔDuct/+ and Pten+/+ mice as controls. We also generated KrasG12D;PtenΔDuct/ΔDuct and KrasG12D;PtenΔDuct/+ mice. Pancreata were collected when mice were 28 weeks to 14.5 months old and analyzed by histology, immunohistochemistry, and electron microscopy. We performed multiplexed droplet digital polymerase chain reaction to detect spontaneous Kras mutations in PtenΔDuct/ΔDuct mice and study the effects of Ras pathway activation on initiation and progression of IPMNs. We obtained 2 pancreatic sections from a patient with an invasive pancreatobiliary IPMN and analyzed the regions with and without the invasive IPMN (control tissue) by immunohistochemistry. RESULTS: Mice with ductal cell-specific disruption of Pten but not control mice developed sporadic, macroscopic, intraductal papillary lesions with histologic and molecular features of human IPMNs. PtenΔDuct/ΔDuct mice developed IPMNs of several subtypes. In PtenΔDuct/ΔDuct mice, 31.5% of IPMNs became invasive; invasion was associated with spontaneous mutations in Kras. KrasG12D;PtenΔDuct/ΔDuct mice all developed invasive IPMNs within 1 month. In KrasG12D;PtenΔDuct/+ mice, 70% developed IPMN, predominately of the pancreatobiliary subtype, and 63.3% developed PDAC. In all models, IPMNs and PDAC expressed the duct-specific lineage tracing marker yellow fluorescent protein. In immunohistochemical analyses, we found that the invasive human pancreatobiliary IPMN tissue had lower levels of PTEN and increased levels of phosphorylated (activated) ERK compared with healthy pancreatic tissue. CONCLUSIONS: In analyses of mice with ductal cell-specific disruption of Pten, with or without activated Kras, we found evidence for a ductal cell origin of IPMNs. We also showed that PTEN loss and activated Kras have synergistic effects in promoting development of IPMN and progression to PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Cell Transformation, Neoplastic/metabolism , Neoplasms, Cystic, Mucinous, and Serous/enzymology , PTEN Phosphohydrolase/deficiency , Pancreatic Ducts/enzymology , Pancreatic Neoplasms/enzymology , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Lineage , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Disease Progression , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neoplasm Invasiveness , Neoplasms, Cystic, Mucinous, and Serous/genetics , Neoplasms, Cystic, Mucinous, and Serous/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Pancreatic Ducts/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phenotype , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction , Time Factors
17.
Endocr Relat Cancer ; 25(1): 11-19, 2018 01.
Article in English | MEDLINE | ID: mdl-29018055

ABSTRACT

Physical activity is associated with a lower risk of disease recurrence among colon cancer survivors. The pathways through which physical activity may alter disease outcomes are unknown, but may include changes in metabolic growth factors, such as insulin. Between January 2015 and August 2015, 39 stage I-III colon cancer survivors were randomized to one of the three groups: usual care control, 150 min/week of aerobic exercise (low-dose) and 300 min/week of aerobic exercise (high-dose) for six months. The pre-specified key metabolic growth factor outcome was fasting insulin. Insulin resistance was quantified using the homeostatic model assessment. Mean age was 56.5 ± 10.0 years, 51% had stage III disease, 72% were treated with chemotherapy and the mean time since finishing treatment was 10.9 ± 6.1 months. Over six months, the low-dose group completed 141.5 ± 9.9 min/week of aerobic exercise, and the high-dose group completed 247.2 ± 10.7 min/week of aerobic exercise. Fasting insulin concentrations decreased 7.4 ± 9.4 pmol/L in the control group, 28.0 ± 8.3 pmol/L in the low-dose group and 20.7 ± 9.3 pmol/L in the high-dose group (nonlinear Ptrend = 0.042). Insulin resistance decreased 0.11 ± 0.20 in the control group, 0.63 ± 0.17 in the low-dose group and 0.43 ± 0.19 in the high-dose group (nonlinear Ptrend = 0.012). Aerobic exercise reduces insulin concentrations and insulin resistance among patients with stage I-III colon cancer. Prescribing 150 min/week of aerobic exercise may be sufficient for reducing insulin concentrations and insulin resistance, which may partially mediate the relationship between physical activity and colon cancer prognosis.


Subject(s)
Cancer Survivors , Colonic Neoplasms/blood , Colonic Neoplasms/therapy , Exercise , Insulin/blood , Adult , Aged , Aged, 80 and over , Body Composition , Colonic Neoplasms/physiopathology , Female , Follow-Up Studies , Humans , Insulin Resistance , Male , Middle Aged , Prognosis
18.
Clin Colorectal Cancer ; 17(1): 32-40, 2018 03.
Article in English | MEDLINE | ID: mdl-28669606

ABSTRACT

BACKGROUND: Observational studies suggest that higher volumes of physical activity are associated with a lower risk of disease recurrence among survivors of colon cancer. However, the feasibility and safety of prescribing higher volumes of physical activity to survivors of colon cancer are unknown. Furthermore, the pathways through which exercise may reduce disease recurrence are unknown. PATIENTS AND METHODS: Survivors of stage I to III colon cancer were randomized to usual-care control, 150 minutes per week of aerobic exercise (low-dose), or 300 minutes per week of aerobic exercise (high-dose). Changes in soluble intercellular adhesion molecule-1 and vascular adhesion molecule-1 prognostic biomarkers were examined. RESULTS: From January 2015 to February 2016, 39 patients were enrolled (n = 13 usual-care control; n = 14 low-dose; n = 12 high-dose), and 38 participants completed the study (97% follow-up). Over 6 months, the low-dose group completed 142 minutes per week (92.8% adherence), and the high-dose group completed 247 minutes per week (89.0% adherence) of exercise. Compared with the control group, changes in soluble intercellular adhesion molecule-1 were -134.9 ng/mL (95% confidence interval, -238.1 to -31.6 ng/mL) in the low-dose group and -114.8 ng/mL (95% confidence interval, -222.5 to -7.1 ng/mL) in the high-dose group (linear Ptrend = .023; nonlinear Ptrend = .044). No changes were observed for soluable vascular adhesion molecule-1 (linear Ptrend = .791; nonlinear Ptrend = .604). Non-serious adverse events occurred at similar rates among randomized groups. No serious adverse events occurred. CONCLUSION: Higher volumes of moderate-intensity aerobic exercise, up to 300 minutes per week, are feasible, safe, and elicit favorable changes in prognostic biomarkers among patients recently treated for stage I to III colon cancer. These data can be used to guide clinical recommendations for patients, and inform future trials.


Subject(s)
Cancer Survivors , Colonic Neoplasms , Exercise Therapy/methods , Adult , Aged , Amine Oxidase (Copper-Containing)/blood , Biomarkers, Tumor/analysis , Cell Adhesion Molecules/blood , Exercise , Female , Humans , Intercellular Adhesion Molecule-1/blood , Male , Middle Aged , Neoplasm Recurrence, Local/prevention & control
19.
Nat Commun ; 8(1): 1728, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29170413

ABSTRACT

Intratumoral phenotypic heterogeneity has been described in many tumor types, where it can contribute to drug resistance and disease recurrence. We analyzed ductal and neuroendocrine markers in pancreatic ductal adenocarcinoma, revealing heterogeneous expression of the neuroendocrine marker Synaptophysin within ductal lesions. Higher percentages of Cytokeratin-Synaptophysin dual positive tumor cells correlate with shortened disease-free survival. We observe similar lineage marker heterogeneity in mouse models of pancreatic ductal adenocarcinoma, where lineage tracing indicates that Cytokeratin-Synaptophysin dual positive cells arise from the exocrine compartment. Mechanistically, MYC binding is enriched at neuroendocrine genes in mouse tumor cells and loss of MYC reduces ductal-neuroendocrine lineage heterogeneity, while deregulated MYC expression in KRAS mutant mice increases this phenotype. Neuroendocrine marker expression is associated with chemoresistance and reducing MYC levels decreases gemcitabine-induced neuroendocrine marker expression and increases chemosensitivity. Altogether, we demonstrate that MYC facilitates ductal-neuroendocrine lineage plasticity in pancreatic ductal adenocarcinoma, contributing to poor survival and chemoresistance.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/metabolism , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Carcinoma, Pancreatic Ductal/drug therapy , Cell Differentiation , Cell Line, Tumor , Cell Lineage , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Drug Resistance, Neoplasm , Female , Heterografts , Humans , Keratins/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Pancreatic Neoplasms/drug therapy , Prognosis , Synaptophysin/metabolism , Gemcitabine
20.
Br J Cancer ; 117(11): 1614-1620, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-28934762

ABSTRACT

BACKGROUND: Physical activity is associated with a lower risk of disease recurrence among colon cancer survivors. Excess visceral adipose tissue is associated with a higher risk of disease recurrence among colon cancer survivors. The pathways through which physical activity may alter disease outcomes are unknown, but may be mediated by changes in visceral adipose tissue. METHODS: Thirty-nine stage I-III colon cancer survivors were randomised to one of three groups: usual-care control, 150 min wk-1 of aerobic exercise (low dose) and 300 min wk-1 of aerobic exercise (high dose) for 6 months. The prespecified key body composition outcome was visceral adipose tissue quantified using dual energy X-ray absorptiometry. RESULTS: Exercise reduced visceral adipose tissue in dose-response fashion (Ptrend=0.008). Compared with the control group, the low- and high-dose exercise groups lost 9.5 cm2 (95% CI: -22.4, 3.5) and 13.6 cm2 (95% CI: -27.0, -0.1) in visceral adipose tissue, respectively. Each 60 min wk-1 increase in exercise predicted a 2.7 cm2 (95% CI: -5.4, -0.1) reduction in visceral adipose tissue. CONCLUSIONS: Aerobic exercise reduces visceral adipose tissue in dose-response fashion among patients with stage I-III colon cancer. Visceral adipose tissue may be a mechanism through which exercise reduces the risk of disease recurrence among colon cancer survivors.


Subject(s)
Body Composition , Cancer Survivors , Colonic Neoplasms/metabolism , Colonic Neoplasms/mortality , Exercise , Adult , Aged , Female , Humans , Intra-Abdominal Fat/metabolism , Male , Middle Aged , Neoplasm Recurrence, Local/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...