Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 96
2.
Cell Mol Gastroenterol Hepatol ; 17(5): 769-784, 2024.
Article En | MEDLINE | ID: mdl-38296052

BACKGROUND & AIMS: Barrett's esophagus is the precursor of esophageal dysplasia and esophageal adenocarcinoma. CDKN2A-p16 deletions were reported in 34%-74% of patients with Barrett's esophagus who progressed to dysplasia and esophageal adenocarcinoma, suggesting that p16 loss may drive neoplastic progression. KRAS activation frequently occurs in esophageal adenocarcinoma and precancer lesions. LGR5+ stem cells in the squamocolumnar-junction (SCJ) of mouse stomach contribute as Barrett's esophagus progenitors. We aimed to determine the functional effects of p16 loss and KRAS activation in Barrett's-like metaplasia and dysplasia development. METHODS: We established mouse models with conditional knockout of CDKN2A-p16 (p16KO) and/or activated KRASG12D expression targeting SCJ LGR5+ cells in interleukin 1b transgenic mice and characterized histologic alterations (mucous-gland hyperplasia/metaplasia, inflammation, and dysplasia) in mouse SCJ. Gene expression was determined by microarray, RNA sequencing, and immunohistochemistry of SCJ tissues and cultured 3-dimensional organoids. RESULTS: p16KO mice exhibited increased mucous-gland hyperplasia/metaplasia versus control mice (P = .0051). Combined p16KO+KRASG12D resulted in more frequent dysplasia and higher dysplasia scores (P = .0036), with 82% of p16KO+KRASG12D mice developing high-grade dysplasia. SCJ transcriptome analysis showed several activated pathways in p16KO versus control mice (apoptosis, tumor necrosis factor-α/nuclear factor-kB, proteasome degradation, p53 signaling, MAPK, KRAS, and G1-to-S transition). CONCLUSIONS: p16 deletion in LGR5+ cell precursors triggers increased SCJ mucous-gland hyperplasia/metaplasia. KRASG12D synergizes with p16 deletion resulting in higher grades of SCJ glandular dysplasia, mimicking Barrett's high-grade dysplasia. These genetically modified mouse models establish a functional role of p16 and activated KRAS in the progression of Barrett's-like lesions to dysplasia in mice, representing an in vivo model of esophageal adenocarcinoma precancer. Derived 3-dimensional organoid models further provide in vitro modeling opportunities of esophageal precancer stages.


Adenocarcinoma , Barrett Esophagus , Esophageal Neoplasms , Precancerous Conditions , Humans , Mice , Animals , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Hyperplasia , Precancerous Conditions/pathology , Adenocarcinoma/pathology , Metaplasia/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism
3.
J Clin Invest ; 133(3)2023 02 01.
Article En | MEDLINE | ID: mdl-36445781

The absence of IFN-γ receptor (IFN-γR) or STAT1 signaling in donor cells has been shown to result in reduced induction of acute graft-versus-host disease (GVHD). In this study, we unexpectedly observed increased activation and expansion of donor lymphocytes in both lymphohematopoietic organs and GVHD target tissues of IFN-γR/STAT1-deficient recipient mice, leading to rapid mortality following the induction of GVHD. LPS-matured, BM-derived Ifngr1-/- Stat1-/- DCs (BMDCs) were more potent allogeneic stimulators and expressed increased levels of MHC II and costimulatory molecules. Similar effects were observed in human antigen-presenting cells (APCs) with knockdown of Stat1 by CRISPR/Cas9 and treatment with a JAK1/2 inhibitor. Furthermore, we demonstrated that the absence of IFN-γR/STAT1 signaling in hematopoietic APCs impaired the presentation of exogenous antigens, while promoting the presentation of endogenous antigens. Thus, the indirect presentation of host antigens to donor lymphocytes was defective in IFN-γR/STAT1-deficient, donor-derived APCs in fully donor chimeric mice. The differential effects of IFN-γR/STAT1 signaling on endogenous and exogenous antigen presentation could provide further insight into the roles of the IFN-γ/STAT1 signaling pathway in the pathogenesis of GVHD, organ rejection, and autoimmune diseases.


Antigen-Presenting Cells , Graft vs Host Disease , Mice , Humans , Animals , Receptors, Interferon/genetics , Graft vs Host Disease/genetics , Signal Transduction , Bone Marrow Transplantation/adverse effects , Mice, Inbred C57BL , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Interferon gamma Receptor
4.
Arch Pathol Lab Med ; 146(10): 1194-1210, 2022 10 01.
Article En | MEDLINE | ID: mdl-35920830

CONTEXT.­: The US Food and Drug Administration (FDA) approved immune checkpoint inhibitor therapy for patients with advanced solid tumors that have DNA mismatch repair defects or high levels of microsatellite instability; however, the FDA provided no guidance on which specific clinical assays should be used to determine mismatch repair status. OBJECTIVE.­: To develop an evidence-based guideline to identify the optimal clinical laboratory test to identify defects in DNA mismatch repair in patients with solid tumor malignancies who are being considered for immune checkpoint inhibitor therapy. DESIGN.­: The College of American Pathologists convened an expert panel to perform a systematic review of the literature and develop recommendations. Using the National Academy of Medicine-endorsed Grading of Recommendations Assessment, Development and Evaluation approach, the recommendations were derived from available evidence, strength of that evidence, open comment feedback, and expert panel consensus. Mismatch repair immunohistochemistry, microsatellite instability derived from both polymerase chain reaction and next-generation sequencing, and tumor mutation burden derived from large panel next-generation sequencing were within scope. RESULTS.­: Six recommendations and 3 good practice statements were developed. More evidence and evidence of higher quality were identified for colorectal cancer and other cancers of the gastrointestinal (GI) tract than for cancers arising outside the GI tract. CONCLUSIONS.­: An optimal assay depends on cancer type. For most cancer types outside of the GI tract and the endometrium, there was insufficient published evidence to recommend a specific clinical assay. Absent published evidence, immunohistochemistry is an acceptable approach readily available in most clinical laboratories.


Colorectal Neoplasms , Microsatellite Instability , Female , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Mismatch Repair/genetics , Immune Checkpoint Inhibitors , Pathologists , Pathology, Molecular/methods , Systematic Reviews as Topic
5.
J Clin Pathol ; 75(9): 593-597, 2022 Sep.
Article En | MEDLINE | ID: mdl-33846218

OBJECTIVES: Misplaced epithelium in adenomas can occasionally be difficult to distinguish from invasive adenocarcinoma. We evaluated interobserver variability in the assessment of left-sided colon polypectomies for pseudoinvasion versus invasive adenocarcinoma and further investigated relevant histological findings. METHODS: 28 consecutive left-sided colon polyps with the keywords "pseudoinvasion", "epithelial misplacement", "herniation", "prolapse" or "invasive adenocarcinoma" were collected from 28 patients and reviewed by eight gastrointestinal pathologists. Participants assessed stromal hemosiderin, lamina propria/eosinophils surrounding glands, desmoplasia, high grade dysplasia/intramucosal adenocarcinoma and margin status and rendered a diagnosis of pseudoinvasion, invasive adenocarcinoma, or both. RESULTS: Agreement among pathologists was substantial for desmoplasia (κ=0.70), high grade dysplasia/intramucosal adenocarcinoma (κ=0.66), invasive adenocarcinoma (κ=0.63) and adenocarcinoma at the margin (κ=0.65). There was moderate agreement for hemosiderin in stroma (κ=0.53) and prolapse/pseudoinvasion (κ=0.50). Agreement was low for lamina propria/eosinophils around glands (κ=0.12). For invasive adenocarcinoma, seven or more pathologists agreed in 24 of 28 cases (86%), and there was perfect agreement in 19/28 cases (68%). For pseudoinvasion, seven or more pathologists agreed in 19 of 28 cases (68%), and there was perfect agreement in 16/28 cases (57%). CONCLUSION: Moderate to substantial, though imperfect, agreement was achieved in the distinction of pseudoinvasion from invasive carcinoma.


Adenocarcinoma , Carcinoma , Colorectal Neoplasms , Adenocarcinoma/pathology , Hemosiderin , Humans , Hyperplasia , Observer Variation
7.
Cancer Prev Res (Phila) ; 14(6): 675-682, 2021 06.
Article En | MEDLINE | ID: mdl-33782049

Hypergastrinemia has been associated with high-grade dysplasia and adenocarcinoma in patients with Barrett's esophagus, and experimental studies suggest proinflammatory and proneoplastic effects of gastrin on Barrett's esophagus. This is of potential concern, as patients with Barrett's esophagus are treated with medications that suppress gastric acid production, resulting in increased physiologic levels of gastrin. We aimed to determine whether treatment with the novel gastrin/CCK2 receptor antagonist netazepide reduces expression of markers associated with inflammation and neoplasia in Barrett's esophagus. This was a randomized, double-blind, placebo-controlled trial of netazepide in patients with Barrett's esophagus without dysplasia. Subjects were treated for 12 weeks, with endoscopic assessment at baseline and at end of treatment. The primary outcome was within-individual change in cellular proliferation as assessed by Ki67. Secondary analyses included changes in gene expression, assessed by RNA-sequencing, and safety and tolerability. A total of 20 subjects completed the study and were included in the analyses. There was no difference between arms in mean change in cellular proliferation (netazepide: +35.6 Ki67+ cells/mm2, SD 620.7; placebo: +307.8 Ki67+ cells/mm2, SD 640.3; P = 0.35). Netazepide treatment resulted in increased expression of genes related to gastric phenotype (TFF2, MUC5B) and certain cancer-associated markers (REG3A, PAX9, MUC1), and decreased expression of intestinal markers MUC2, FABP1, FABP2, and CDX1 No serious adverse events related to study drug occurred. The gastrin/CCK2 receptor antagonist netazepide did not reduce cellular proliferation in patients with nondysplastic Barrett's esophagus. Further research should focus on the biological effects of gastrin in Barrett's esophagus.Prevention Relevance: Treatment of patients with Barrett's esophagus with a gastrin/CCK2 receptor antagonist did not have obvious chemopreventive effects.


Adenocarcinoma/prevention & control , Barrett Esophagus/drug therapy , Benzodiazepinones/administration & dosage , Esophageal Neoplasms/prevention & control , Phenylurea Compounds/administration & dosage , Adenocarcinoma/diagnosis , Adenocarcinoma/pathology , Aged , Barrett Esophagus/pathology , Benzodiazepinones/adverse effects , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Double-Blind Method , Esophageal Mucosa/diagnostic imaging , Esophageal Mucosa/drug effects , Esophageal Mucosa/pathology , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/pathology , Esophagoscopy , Female , Gastrins/antagonists & inhibitors , Humans , Male , Middle Aged , Phenylurea Compounds/adverse effects , Receptor, Cholecystokinin B/antagonists & inhibitors
8.
Cancer Res ; 80(16): 3265-3278, 2020 08 15.
Article En | MEDLINE | ID: mdl-32540961

Activation of oncogenic KRAS is the most common driving event in lung adenocarcinoma development. Despite the existing rationale for targeting activated KRAS and its downstream effectors, the failure of clinical trials to date indicates that the mechanism of KRAS-driven malignancy remains poorly understood. Here we report that histone deacetylase 10 (HDAC10) might function as a putative tumor suppressor in mice carrying a spontaneously activated oncogenic Kras allele. Hdac10 deletion accelerated KRAS-driven early-onset lung adenocarcinomas, increased macrophage infiltration in the tumor microenvironment, and shortened survival time in mice. Highly tumorigenic and stem-like lung adenocarcinoma cells were increased in Hdac10-deleted tumors compared with Hdac10 wild-type tumors. HDAC10 regulated the stem-like properties of KRAS-expressing tumor cells by targeting SOX9. Expression of SOX9 was significantly increased in Hdac10-deleted tumor cells and depletion of SOX9 in Hdac10 knockout (KO) lung adenocarcinoma cells inhibited growth of tumorspheres. The genes associated with TGFß pathway were enriched in Hdac10 KO tumor cells, and activation of TGFß signaling contributed to SOX9 induction in Hdac10 KO lung adenocarcinoma cells. Overall, our study evaluates the functions and mechanisms of action of HDAC10 in lung carcinogenesis that will inform the rationale for targeting its related regulatory signaling as an anticancer strategy. SIGNIFICANCE: These findings linking HDAC10 and lung tumorigenesis identify potential novel strategies for targeting HDAC10 as a treatment for lung cancer.


Adenocarcinoma of Lung/genetics , Genes, ras , Histone Deacetylases/genetics , Lung Neoplasms/genetics , Transcriptional Activation , Tumor Suppressor Proteins/genetics , Adenocarcinoma of Lung/mortality , Adenocarcinoma of Lung/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line, Tumor , Disease Models, Animal , Gene Deletion , Germ-Line Mutation , Histone Deacetylases/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Macrophages , Mice , Neoplastic Stem Cells , SOX9 Transcription Factor/metabolism , Sequence Analysis, RNA , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Microenvironment , Tumor Suppressor Proteins/metabolism
9.
Gastroenterology ; 159(2): 575-590, 2020 08.
Article En | MEDLINE | ID: mdl-32325086

BACKGROUND & AIMS: Studies are needed to determine the mechanism by which Barrett's esophagus (BE) progresses to esophageal adenocarcinoma (EAC). Notch signaling maintains stem cells in the gastrointestinal tract and is dysregulated during carcinogenesis. We explored the relationship between Notch signaling and goblet cell maturation, a feature of BE, during EAC pathogenesis. METHODS: We measured goblet cell density and levels of Notch messenger RNAs in BE tissues from 164 patients, with and without dysplasia or EAC, enrolled in a multicenter study. We analyzed the effects of conditional expression of an activated form of NOTCH2 (pL2.Lgr5.N2IC), conditional deletion of NOTCH2 (pL2.Lgr5.N2fl/fl), or loss of nuclear factor κB (NF-κB) (pL2.Lgr5.p65fl/fl), in Lgr5+ (progenitor) cells in L2-IL1B mice (which overexpress interleukin 1 beta in esophagus and squamous forestomach and are used as a model of BE). We collected esophageal and stomach tissues and performed histology, immunohistochemistry, flow cytometry, transcriptome, and real-time polymerase chain reaction analyses. Cardia and forestomach tissues from mice were cultured as organoids and incubated with inhibitors of Notch or NF-kB. RESULTS: Progression of BE to EAC was associated with a significant reduction in goblet cell density comparing nondysplastic regions of tissues from patients; there was an inverse correlation between goblet cell density and levels of NOTCH3 and JAG2 messenger RNA. In mice, expression of the activated intracellular form of NOTCH2 in Lgr5+ cells reduced goblet-like cell maturation, increased crypt fission, and accelerated the development of tumors in the squamocolumnar junction. Mice with deletion of NOTCH2 from Lgr5+ cells had increased maturation of goblet-like cells, reduced crypt fission, and developed fewer tumors. Esophageal tissues from in pL2.Lgr5.N2IC mice had increased levels of RelA (which encodes the p65 unit of NF-κB) compared to tissues from L2-IL1B mice, and we found evidence of increased NF-κB activity in Lgr5+ cells. Esophageal tissues from pL2.Lgr5.p65fl/fl mice had lower inflammation and metaplasia scores than pL2.Lgr5.N2IC mice. In organoids derived from pL2-IL1B mice, the NF-κB inhibitor JSH-23 reduced cell survival and proliferation. CONCLUSIONS: Notch signaling contributes to activation of NF-κB and regulates differentiation of gastric cardia progenitor cells in a mouse model of BE. In human esophageal tissues, progression of BE to EAC was associated with reduced goblet cell density and increased levels of Notch expression. Strategies to block this pathway might be developed to prevent EAC in patients with BE.


Adenocarcinoma/pathology , Barrett Esophagus/pathology , Carcinogenesis/pathology , Esophageal Neoplasms/pathology , Goblet Cells/pathology , Receptors, Notch/metabolism , Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Aged , Animals , Barrett Esophagus/diagnosis , Barrett Esophagus/genetics , Biopsy , Carcinogenesis/genetics , Cell Differentiation/genetics , Cross-Sectional Studies , Disease Models, Animal , Disease Progression , Esophageal Mucosa/cytology , Esophageal Mucosa/diagnostic imaging , Esophageal Mucosa/pathology , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/genetics , Esophagoscopy , Female , Gastric Mucosa/cytology , Gastric Mucosa/pathology , Humans , Male , Mice , Mice, Transgenic , Middle Aged , NF-kappa B/metabolism , Prospective Studies , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptors, Notch/genetics , Signal Transduction
10.
Cell Mol Gastroenterol Hepatol ; 10(2): 434-449.e1, 2020.
Article En | MEDLINE | ID: mdl-32330731

BACKGROUND & AIMS: Enterochromaffin-like (ECL) cells in the stomach express gastrin/cholecystokinin 2 receptor CCK2R and are known to expand under hypergastrinemia, but whether this results from expansion of existing ECL cells or increased production from progenitors has not been clarified. METHODS: We used mice with green fluorescent protein fluorescent reporter expression in ECL cells (histidine decarboxylase [Hdc]-green fluorescent protein), as well as Cck2r- and Hdc-driven Tamoxifen inducible recombinase Cre (Cck2r-CreERT2, Hdc-CreERT2) mice combined with Rosa26Sor-tdTomato (R26-tdTomato) mice, and studied their expression and cell fate in the gastric corpus by using models of hypergastrinemia (gastrin infusion, omeprazole treatment). RESULTS: Hdc-GFP marked the majority of ECL cells, located in the lower third of the gastric glands. Hypergastrinemia led to expansion of ECL cells that was not restricted to the gland base, and promoted cellular proliferation (Ki67) in the gastric isthmus but not in basal ECL cells. Cck2r-CreERT2 mice marked most ECL cells, as well as scattered cell types located higher up in the glands, whose number was increased during hypergastrinemia. Cck2r-CreERT2+ isthmus progenitors, but not Hdc+ mature ECL cells, were the source of ECL cell hyperplasia during hypergastrinemia and could grow as 3-dimensional spheroids in vitro. Moreover, gastrin treatment in vitro promoted sphere formation from sorted Cck2r+Hdc- cells, and increased chromogranin A and phosphorylated- extracellular signal-regulated kinase expression in CCK2R-derived organoids. Gastrin activates extracellular signal-regulated kinase pathways in vivo and in vitro, and treatment with the Mitogen-activated protein kinase kinase 1 inhibitor U0126 blocked hypergastrinemia-mediated changes, including CCK2R-derived ECL cell hyperplasia in vivo as well as sphere formation and chromogranin A expression in vitro. CONCLUSIONS: We show here that hypergastrinemia induces ECL cell hyperplasia that is derived primarily from CCK2R+ progenitors in the corpus. Gastrin-dependent function of CCK2R+ progenitors is regulated by the extracellular signal-regulated kinase pathway.


Enterochromaffin-like Cells/pathology , Gastric Mucosa/pathology , Gastrins/blood , Animals , Disease Models, Animal , Enterochromaffin-like Cells/metabolism , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Gastrins/metabolism , Humans , Hyperplasia/blood , Hyperplasia/pathology , MAP Kinase Signaling System , Mice , Receptor, Cholecystokinin B/metabolism , Stem Cells/pathology
11.
Cell Stem Cell ; 26(5): 739-754.e8, 2020 05 07.
Article En | MEDLINE | ID: mdl-32142681

Cancer is believed to arise from stem cells, but mechanisms that limit the acquisition of mutations and tumor development have not been well defined. We show that a +4 stem cell (SC) in the gastric antrum, marked by expression of Cck2r (a GPCR) and Delta-like ligand 1 (DLL1), is a label-retaining cell that undergoes predominant asymmetric cell division. This +4 antral SC is Notch1low/ Numb+ and repressed by signaling from gastrin-expressing endocrine (G) cells. Chemical carcinogenesis of the stomach is associated with loss of G cells, increased symmetric stem cell division, glandular fission, and more rapid stem cell lineage tracing, a process that can be suppressed by exogenous gastrin treatment. This hormonal suppression is associated with a marked reduction in gastric cancer mutational load, as revealed by exomic sequencing. Taken together, our results show that gastric tumorigenesis is associated with increased symmetric cell division that facilitates mutation and is suppressed by GPCR signaling.


Carcinogenesis , Stem Cells , Cell Division , Humans , Signal Transduction , Stomach
12.
J Immunother Cancer ; 8(1)2020 01.
Article En | MEDLINE | ID: mdl-31949040

BACKGROUND: Quality measures are important because they can help improve and standardize the delivery of cancer care among healthcare providers and across tumor types. In an environment characterized by a rapidly shifting immunotherapeutic landscape and lack of associated long-term outcome data, defining quality measures for cancer immunotherapy is a high priority yet fraught with many challenges. METHODS: Thus, the Society for Immunotherapy of Cancer convened a multistakeholder expert panel to, first, identify the current gaps in measures of quality cancer care delivery as it relates to immunotherapy and to, second, advance priority concepts surrounding quality measures that could be developed and broadly adopted by the field. RESULTS: After reviewing the existing quality measure landscape employed for immunotherapeutic-based cancer care, the expert panel identified four relevant National Quality Strategy domains (patient safety, person and family-centered care, care coordination and communication, appropriate treatment selection) with significant gaps in immunotherapy-based quality cancer care delivery. Furthermore, these domains offer opportunities for the development of quality measures as they relate to cancer immunotherapy. These four quality measure concepts are presented in this consensus statement. CONCLUSIONS: This work represents a first step toward defining and standardizing quality delivery of cancer immunotherapy in order to realize its optimal application and benefit for patients.


Immunotherapy/methods , Neoplasms/therapy , History, 21st Century , Humans
13.
Oncotarget ; 10(39): 3852-3864, 2019 Jun 11.
Article En | MEDLINE | ID: mdl-31231464

Gastric adenocarcinoma (GC) is a leading cause of cancer-related deaths worldwide. The transcription factor gene Friend Leukemia Integration 1 (FLI1) is methylated and downregulated in human GC tissues. Using human GC samples, we determined which cells downregulate FLI1, when FLI1 downregulation occurs, if FLI1 downregulation correlates with clinical-pathologic characteristics, and whether FLI1 plays a role in invasion and/or proliferation of cultured cells. We analyzed stomach tissues from 98 patients [8 normal mucosa, 8 intestinal metaplasia (IM), 7 dysplasia, 91 GC] by immunohistochemistry for FLI1. Epithelial cells from normal, IM, and low-grade dysplasia (LGD) showed strong nuclear FLI1 staining. GC epithelial cells showed significantly less nuclear FLI1 staining as compared to normal epithelium, IM and LGD (P=1.2×10-5, P=1.4×10-6 and P=0.006, respectively). FLI1 expression did not correlate with tumor stage or differentiation, but was associated with patient survival, depending on tumor differentiation. We tested the functional role of FLI1 by assaying proliferation and invasion in cultured GC cells. Lentiviral-transduced FLI1 overexpression in GC AGS cells inhibited invasion by 73.5% (P = 0.001) and proliferation by 31.5% (P = 0.002), as compared to controls. Our results support a combined role for FLI1 as a suppressor of invasiveness and proliferation in gastric adenocarcinoma, specifically in the transition from pre-cancer lesions and dysplasia to invasive adenocarcinoma, and suggest that FLI1 may be a prognostic biomarker of survival in gastric cancers.

14.
J Clin Pathol ; 72(7): 460-467, 2019 Jul.
Article En | MEDLINE | ID: mdl-31072837

The neurotrophic tyrosine receptor kinase (NTRK) gene family encodes three tropomyosin receptor kinases (TRKA, TRKB, TRKC) that contribute to central and peripheral nervous system development and function. NTRK gene fusions are oncogenic drivers of various adult and paediatric tumours. Several methods have been used to detect NTRK gene fusions including immunohistochemistry, fluorescence in situ hybridisation, reverse transcriptase polymerase chain reaction, and DNA- or RNA-based next-generation sequencing. For patients with TRK fusion cancer, TRK inhibition is an important therapeutic target. Following the FDA approval of the selective TRK inhibitor, larotrectinib, as well as the ongoing development of multi-kinase inhibitors with activity in TRK fusion cancer, testing for NTRK gene fusions should become part of the standard diagnostic process. In this review we discuss the biology of NTRK gene fusions, and we present a testing algorithm to aid detection of these gene fusions in clinical practice and guide treatment decisions.


Neoplasms/genetics , Nerve Growth Factors/genetics , Oncogene Proteins, Fusion/genetics , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Receptor Protein-Tyrosine Kinases/genetics , Algorithms , Humans , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Neoplasms/drug therapy , Neoplasms/enzymology , Nerve Growth Factors/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/genetics , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/genetics , Receptor, trkC/antagonists & inhibitors , Receptor, trkC/genetics
15.
Int J Cancer ; 145(10): 2754-2766, 2019 11 15.
Article En | MEDLINE | ID: mdl-31001805

The main risk factor for esophageal dysplasia and adenocarcinoma (DAC) is Barrett's esophagus (BE), characterized by intestinal metaplasia. The critical genomic mechanisms that lead to progression of nondysplastic BE to DAC remain poorly understood and require analyses of longitudinal patient cohorts and high-resolution assays. We tested BE tissues from 74 patients, including 42 nonprogressors from two separate groups of 21 patients each and 32 progressors (16 in a longitudinal cohort before DAC/preprogression-BE and 16 with temporally concurrent but spatially separate DAC/concurrent-BE). We interrogated genome-wide somatic copy number alterations (SCNAs) at the exon level with high-resolution SNP arrays in DNA from formalin-fixed samples histologically confirmed as nondysplastic BE. The most frequent abnormalities were SCNAs involving FHIT exon 5, CDKN2A/B or both in 88% longitudinal BE progressors to DAC vs. 24% in both nonprogressor groups (p = 0.0004). Deletions in other genomic regions were found in 56% of preprogression-BE but only in one nonprogressor-BE (p = 0.0004). SCNAs involving FHIT exon 5 and CDKN2A/B were also frequently detected in BE temporally concurrent with DAC. TP53 losses were detected in concurrent-BE but not earlier in preprogression-BE tissues of patients who developed DAC. CDKN2A/p16 immunohistochemistry showed significant loss of expression in BE of progressors vs. nonprogressors, supporting the genomic data. Our data suggest a role for CDKN2A/B and FHIT in early progression of BE to dysplasia and adenocarcinoma that warrants future mechanistic research. Alterations in CDKN2A/B and FHIT by high-resolution assays may serve as biomarkers of increased risk of progression to DAC when detected in BE tissues.


Adenocarcinoma/pathology , Barrett Esophagus/genetics , Biomarkers, Tumor/genetics , Esophageal Mucosa/pathology , Esophageal Neoplasms/pathology , Precancerous Conditions/genetics , Acid Anhydride Hydrolases/genetics , Adult , Aged , Barrett Esophagus/pathology , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Copy Number Variations , Disease Progression , Exons/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Longitudinal Studies , Male , Middle Aged , Neoplasm Proteins/genetics , Polymorphism, Single Nucleotide , Precancerous Conditions/pathology , Tumor Suppressor Protein p53/genetics
16.
Gut ; 68(6): 1034-1043, 2019 06.
Article En | MEDLINE | ID: mdl-30658994

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDA) has among the highest stromal fractions of any cancer and this has complicated attempts at expression-based molecular classification. The goal of this work is to profile purified samples of human PDA epithelium and stroma and examine their respective contributions to gene expression in bulk PDA samples. DESIGN: We used laser capture microdissection (LCM) and RNA sequencing to profile the expression of 60 matched pairs of human PDA malignant epithelium and stroma samples. We then used these data to train a computational model that allowed us to infer tissue composition and generate virtual compartment-specific expression profiles from bulk gene expression cohorts. RESULTS: Our analysis found significant variation in the tissue composition of pancreatic tumours from different public cohorts. Computational removal of stromal gene expression resulted in the reclassification of some tumours, reconciling functional differences between different cohorts. Furthermore, we established a novel classification signature from a total of 110 purified human PDA stroma samples, finding two groups that differ in the extracellular matrix-associated and immune-associated processes. Lastly, a systematic evaluation of cross-compartment subtypes spanning four patient cohorts indicated partial dependence between epithelial and stromal molecular subtypes. CONCLUSION: Our findings add clarity to the nature and number of molecular subtypes in PDA, expand our understanding of global transcriptional programmes in the stroma and harmonise the results of molecular subtyping efforts across independent cohorts.


Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/surgery , Carcinoma, Pancreatic Ductal/surgery , Case-Control Studies , Computer Simulation , Extracellular Matrix/pathology , Gene Expression Profiling , Humans , Microdissection , Pancreatic Neoplasms/surgery , Sensitivity and Specificity
17.
Appl Immunohistochem Mol Morphol ; 27(9): 694-698, 2019 10.
Article En | MEDLINE | ID: mdl-30499817

BACKGROUND: Utilization rates of immunohistochemistry (IHC) for the diagnosis of Helicobacter pylori infection may vary by laboratory and/or pathologists. IHC for H. pylori is not performed routinely in our practice. Instead, it is used in selected cases at the pathologists' discretion (and according to their specific criteria). The purpose of this study was to determine if IHC utilization rates correlated with rates of detecting H. pylori infection. MATERIALS AND METHODS: We searched our records and investigated all gastric biopsies for 1 calendar year. H. pylori diagnostic rate and IHC utilization rate was calculated for each pathologist. RESULTS: Overall, the rate of diagnosis was 12.1% and the IHC utilization rate was 45.2%. Individual pathologists had H. pylori diagnostic rates ranging from 3.6% to 34.1% (median: 11.1%) and IHC utilization ranging from 17.1% to 95.2% (median: 42.2%). The rate of detection of H. pylori infection among pathologists showed no significant correlation with rates of IHC utilization (Pearson coefficient=0.121). CONCLUSIONS: Increasing use of IHC is not independently associated with the diagnostic rate of infection. Ultimately, if we assume that the case mix was similar for each pathologist, it suggests that more liberal criteria to order IHC does not result in more infections diagnosed.


Gastric Mucosa/pathology , Helicobacter Infections/diagnosis , Helicobacter pylori/physiology , Immunohistochemistry/statistics & numerical data , Procedures and Techniques Utilization/statistics & numerical data , Adult , Biopsy , Diagnostic Tests, Routine , Female , Helicobacter Infections/epidemiology , Humans , Male , Middle Aged , Staining and Labeling , United States/epidemiology
18.
Nat Genet ; 50(7): 979-989, 2018 07.
Article En | MEDLINE | ID: mdl-29915428

We introduce and validate a new precision oncology framework for the systematic prioritization of drugs targeting mechanistic tumor dependencies in individual patients. Compounds are prioritized on the basis of their ability to invert the concerted activity of master regulator proteins that mechanistically regulate tumor cell state, as assessed from systematic drug perturbation assays. We validated the approach on a cohort of 212 gastroenteropancreatic neuroendocrine tumors (GEP-NETs), a rare malignancy originating in the pancreas and gastrointestinal tract. The analysis identified several master regulator proteins, including key regulators of neuroendocrine lineage progenitor state and immunoevasion, whose role as critical tumor dependencies was experimentally confirmed. Transcriptome analysis of GEP-NET-derived cells, perturbed with a library of 107 compounds, identified the HDAC class I inhibitor entinostat as a potent inhibitor of master regulator activity for 42% of metastatic GEP-NET patients, abrogating tumor growth in vivo. This approach may thus complement current efforts in precision oncology.


Antineoplastic Agents/pharmacology , Neuroendocrine Tumors/drug therapy , Benzamides/pharmacology , Cell Line, Tumor , Cohort Studies , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Intestinal Neoplasms/drug therapy , Intestinal Neoplasms/genetics , Neuroendocrine Tumors/genetics , Pancreas/drug effects , Pancreas/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Precision Medicine/methods , Pyridines/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics
19.
Arch Pathol Lab Med ; 142(4): 516-522, 2018 Apr.
Article En | MEDLINE | ID: mdl-28782986

CONTEXT: - In advanced gastric, esophageal, and gastroesophageal junction adenocarcinomas (GE-GEJ-AC) that overexpress ERBB2 (erb-b2 receptor tyrosine kinase 2 or HER2), anti-HER2 monoclonal antibody therapy confers survival benefit. To select patients for treatment, HER2 expression and gene amplification are evaluated by immunohistochemistry (IHC) and in situ hybridization. OBJECTIVE: - To determine whether GE-GEJ-AC tested for HER2 on biopsy specimens of a primary tumor show different IHC scores and/or HER2 amplification by in situ hybridization in matched resection specimens, potentially changing therapy eligibility. DESIGN: - Immunohistochemistry and silver in situ hybridization were performed in biopsy and/or resection specimens from 100 patients. HER2 testing was performed in matched resection and biopsy specimens of 15 cases to determine whether GE-GEJ-AC with IHC scores of 0, 1+, and 2+ in biopsy and resection specimens had different IHC and silver in situ hybridization results. RESULTS: - The IHC 3+ cases showed HER2 amplification in 4 of 5 cases (80%), and IHC scores of 0, 1+, and 2+ showed 3.5%, 14.3%, and 23.5% HER2 amplification by silver in situ hybridization. Among the 15 paired biopsy and resection specimens, 9 (60%) had at least pT2 stage GE-GEJ-AC with HER2 IHC scores of 0, 1+, or 2+ in the biopsy, and 2 of those 9 cases (22%) had IHC 3+ and HER2 amplification by silver in situ hybridization on the resection specimen. CONCLUSIONS: - Our data suggest that HER2 testing should be repeated on resection specimens of GE-GEJ-AC with HER2 IHC scores of negative (0 and 1+) or equivocal (2+) and in situ hybridization amplification negative biopsy specimen results to evaluate for HER2 heterogeneity when patients are being considered for anti-HER2 therapy.


Adenocarcinoma/diagnosis , Biomarkers, Tumor/analysis , Esophageal Neoplasms/diagnosis , Receptor, ErbB-2/analysis , Stomach Neoplasms/diagnosis , Esophagogastric Junction/pathology , Humans , Immunohistochemistry/methods , In Situ Hybridization
20.
Clin Cancer Res ; 24(5): 1048-1061, 2018 03 01.
Article En | MEDLINE | ID: mdl-29208671

Purpose: Barrett's esophagus represents an early stage in carcinogenesis leading to esophageal adenocarcinoma. Considerable evidence supports a major role for chronic inflammation and diverse chemokine pathways in the development of Barrett's esophagus and esophageal adenocarcinoma.Experimental Design: Here we utilized an IL1ß transgenic mouse model of Barrett's esophagus and esophageal adenocarcinoma and human patient imaging to analyze the importance of CXCR4-expressing cells during esophageal carcinogenesis.Results: IL1ß overexpression induces chronic esophageal inflammation and recapitulates the progression to Barrett's esophagus and esophageal adenocarcinoma. CXCR4 expression is increased in both epithelial and immune cells during disease progression in pL2-IL1ß mice and also elevated in esophageal adenocarcinoma patient biopsy samples. Specific recruitment of CXCR4-positive (CXCR4+) immune cells correlated with dysplasia progression, suggesting that this immune population may be a key contributor to esophageal carcinogenesis. Similarly, with progression to dysplasia, there were increased numbers of CXCR4+ columnar epithelial cells at the squamocolumnar junction (SCJ). These findings were supported by stronger CXCR4-related signal intensity in ex vivo fluorescence imaging and autoradiography with advanced dysplasia. Pilot CXCR4-directed PET/CT imaging studies in patients with esophageal cancer demonstrate the potential utility of CXCR4 imaging for the diagnosis and staging of esophageal cancer.Conclusion: In conclusion, the recruitment of CXCR4+ immune cells and expansion of CXCR4+ epithelial cells in esophageal dysplasia and cancer highlight the potential of CXCR4 as a biomarker and molecular target for diagnostic imaging of the tumor microenvironment in esophageal adenocarcinoma. Clin Cancer Res; 24(5); 1048-61. ©2017 AACR.


Adenocarcinoma/pathology , Barrett Esophagus/pathology , Biomarkers, Tumor/metabolism , Esophageal Neoplasms/pathology , Receptors, CXCR4/metabolism , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/immunology , Aged , Aged, 80 and over , Animals , Barrett Esophagus/diagnostic imaging , Barrett Esophagus/immunology , Biomarkers, Tumor/immunology , Biopsy , Carcinogenesis/pathology , Coordination Complexes/administration & dosage , Disease Models, Animal , Disease Progression , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/immunology , Esophagus/immunology , Esophagus/pathology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice, Transgenic , Middle Aged , Molecular Imaging/methods , Peptides, Cyclic/administration & dosage , Positron Emission Tomography Computed Tomography/methods , Receptors, CXCR4/immunology , Tissue Array Analysis , Tumor Microenvironment/immunology , Up-Regulation
...