318 results on '"Hanash SM"'
Search Results
2. Co-amplification of a novel gene, NAG, with the N-myc gene in neuroblastoma
- Author
-
Wimmer, K, Zhu, XX, Lamb, BJ, Kuick, R, Ambros, PF, Kovar, H, Thoraval, D, Motyka, S, Alberts, JR, and Hanash, SM
- Published
- 1999
- Full Text
- View/download PDF
3. Quantitative analysis of Op18 phosphorylation in childhood acute leukemia
- Author
-
Melhem, R, Hailat, N, Kuick, R, and Hanash, SM
- Published
- 1997
- Full Text
- View/download PDF
4. Q18E mutation of Op18 detected in a gastric cardia tumor inhibits cell differentiation
- Author
-
Chang, CL, primary, Melhem, RF, additional, Beer, DG, additional, Kuick, R, additional, Hinderer, R, additional, and Hanash, SM, additional
- Published
- 1998
- Full Text
- View/download PDF
5. A role for chromosome 1 in colorectal cancer
- Author
-
Hanash, SM, primary
- Published
- 1996
- Full Text
- View/download PDF
6. Emerging molecular biomarkers--blood-based strategies to detect and monitor cancer.
- Author
-
Hanash SM, Baik CS, Kallioniemi O, Hanash, Samir M, Baik, Christina S, and Kallioniemi, Olli
- Abstract
There is an urgent need for blood-based, noninvasive molecular tests to assist in the detection and diagnosis of cancers in a cost-effective manner at an early stage, when curative interventions are still possible. Additionally, blood-based diagnostics can classify tumors into distinct molecular subtypes and monitor disease relapse and response to treatment. Increasingly, biomarker strategies are becoming critical to identify a specific patient subpopulation that is likely to respond to a new therapeutic agent. The improved understanding of the underlying molecular features of common cancers and the availability of a multitude of recently developed technologies to interrogate the genome, transcriptome, proteome and metabolome of tumors and biological fluids have made it possible to develop clinically applicable and cost-effective tests for many common cancers. Overall, the paradigm shift towards personalized and individualized medicine relies heavily on the increased use of diagnostic biomarkers and classifiers to improve diagnosis, management and treatment. International collaborations, involving both the private and public sector will be required to facilitate the development of clinical applications of biomarkers, using rigorous standardized assays. Here, we review the recent technological and scientific advances in this field. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
7. Occurrence of autoantibodies to annexin I, 14-3-3 theta and LAMR1 in prediagnostic lung cancer sera.
- Author
-
Qiu J, Choi G, Li L, Wang H, Pitteri SJ, Pereira-Faca SR, Krasnoselsky AL, Randolph TW, Omenn GS, Edelstein C, Barnett MJ, Thornquist MD, Goodman GE, Brenner DE, Feng Z, Hanash SM, Qiu, Ji, Choi, Gina, Li, Lin, and Wang, Hong
- Published
- 2008
- Full Text
- View/download PDF
8. Proliferating cell nuclear antigen expression in childhood acute leukemia
- Author
-
Keim, D, primary, Hailat, N, additional, Hodge, D, additional, and Hanash, SM, additional
- Published
- 1990
- Full Text
- View/download PDF
9. p53 mutation as a prognostic marker in advanced laryngeal carcinoma. Department of Veterans Affairs Laryngeal Cancer Cooperative Study Group.
- Author
-
Bradford CR, Zhu S, Poore J, Fisher SG, Beals TF, Thoraval D, Hanash SM, Carey TE, and Wolf GT
- Published
- 1997
10. Identification of a cellular polypeptide that distinguishes between acute lymphoblastic leukemia in infants and in older children
- Author
-
Hanash, SM, Kuick, R, Strahler, J, Richardson, B, Reaman, G, Stoolman, L, Hanson, C, Nichols, D, and Tueche, HJ
- Abstract
We analyzed the polypeptide pattern of leukemic cells of infants and older children with acute lymphoblastic leukemia (ALL), using two- dimensional polyacrylamide gel electrophoresis (PAGE). Patterns were analyzed for the occurrence of a previously detected cytosolic polypeptide, designated L3. Quantitative analysis of L3 in 12 infants and 91 older children with non-T ALL indicated lack of expression of polypeptide L3 in leukemic cells of infants which, in most cases, expressed HLA-DR and CD19 and lacked CD10. Quantitative analysis of L3 in relation to cell surface marker expression revealed that L3 was limited in its occurrence to non-T ALL and was not coordinately expressed with any of the surface markers included in the study. Among patients in the HLA-DR-positive, CD19-positive, and CD10-negative group, different levels of polypeptide L3 were observed between infants and older children. These results indicate differences in leukemic cell constituents between infants and older children with ALL and an otherwise similar cell surface marker phenotype.
- Published
- 1989
- Full Text
- View/download PDF
11. A compendium of potential biomarkers of pancreatic cancer.
- Author
-
Harsha HC, Kandasamy K, Ranganathan P, Rani S, Ramabadran S, Gollapudi S, Balakrishnan L, Dwivedi SB, Telikicherla D, Selvan LD, Goel R, Mathivanan S, Marimuthu A, Kashyap M, Vizza RF, Mayer RJ, Decaprio JA, Srivastava S, Hanash SM, and Hruban RH
- Published
- 2009
- Full Text
- View/download PDF
12. Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers
- Author
-
Gloria M. Petersen, Ammar A. Javed, Alison P. Klein, Matthew J. Weiss, Janine Ptak, Nickolas Papadopoulos, Ralph H. Hruban, Peter Gibbs, Peter J. Allen, Martin A. Makary, Marco Dal Molin, Jin He, Cristian Tomasetti, Yuxuan Wang, Natalie Silliman, Lisa Dobbyn, Lu Li, Christopher L. Wolfgang, Jeanne Tie, Christopher J. Thoburn, Bert Vogelstein, Fay Wong, Claudio Doglioni, Kenneth W. Kinzler, Michele T. Yip-Schneider, Randall E. Brand, Maria Popoli, Massimo Falconi, Aatur D. Singhi, Samir M. Hanash, Mark A. Schattner, Anirban Maitra, Seung-Mo Hong, Joshua D. Cohen, Joy Schaefer, Michael Goggins, C. Max Schmidt, Song Cheol Kim, Nita Ahuja, Anne Marie Lennon, Cohen, Jd, Javed, Aa, Thoburn, C, Wong, F, Tie, J, Gibbs, P, Schmidt, Cm, Yip-Schneider, Mt, Allen, Pj, Schattner, M, Brand, Re, Singhi, Ad, Petersen, Gm, Hong, Sm, Kim, Sc, Falconi, M, Doglioni, C, Weiss, Mj, Ahuja, N, He, J, Makary, Ma, Maitra, A, Hanash, Sm, Dal Molin, M, Wang, Y, Li, L, Ptak, J, Dobbyn, L, Schaefer, J, Silliman, N, Popoli, M, Goggins, Mg, Hruban, Rh, Wolfgang, Cl, Klein, Ap, Tomasetti, C, Papadopoulos, N, Kinzler, Kw, Vogelstein, B, and Lennon, Am
- Subjects
Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Pathology ,CA-19-9 Antigen ,Biology ,Gene mutation ,medicine.disease_cause ,Circulating Tumor DNA ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,0302 clinical medicine ,Pancreatic cancer ,Internal medicine ,medicine ,Carcinoma ,Humans ,Liquid biopsy ,Aged ,Multidisciplinary ,Liquid Biopsy ,Cancer ,Middle Aged ,Biological Sciences ,Genes, p53 ,medicine.disease ,Primary tumor ,Pancreatic Neoplasms ,030104 developmental biology ,Case-Control Studies ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Female ,KRAS ,Carcinoma, Pancreatic Ductal - Abstract
The earlier diagnosis of cancer is one of the keys to reducing cancer deaths in the future. Here we describe our efforts to develop a noninvasive blood test for the detection of pancreatic ductal adenocarcinoma. We combined blood tests for KRAS gene mutations with carefully thresholded protein biomarkers to determine whether the combination of these markers was superior to any single marker. The cohort tested included 221 patients with resectable pancreatic ductal adenocarcinomas and 182 control patients without known cancer. KRAS mutations were detected in the plasma of 66 patients (30%), and every mutation found in the plasma was identical to that subsequently found in the patient's primary tumor (100% concordance). The use of KRAS in conjunction with four thresholded protein biomarkers increased the sensitivity to 64%. Only one of the 182 plasma samples from the control cohort was positive for any of the DNA or protein biomarkers (99.5% specificity). This combinatorial approach may prove useful for the earlier detection of many cancer types.
- Published
- 2017
13. Atlas of Metastatic Gastric Cancer Links Ferroptosis to Disease Progression and Immunotherapy Response.
- Author
-
Cheng X, Dai E, Wu J, Flores NM, Chu Y, Wang R, Dang M, Xu Z, Han G, Liu Y, Chatterjee D, Hu C, Ying J, Du Y, Yang L, Guan X, Mo S, Cao X, Pei G, Jiang J, Lu X, Benitez AM, Waters RE, Pizzi MP, Shanbhag N, Fan Y, Peng F, Hanash SM, Calin G, Futreal A, Song S, Yee C, Mazur PK, Qin JJ, Ajani JA, and Wang L
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, Peritoneal Neoplasms secondary, Peritoneal Neoplasms immunology, Peritoneal Neoplasms genetics, Peritoneal Neoplasms therapy, Phospholipid Hydroperoxide Glutathione Peroxidase metabolism, Phospholipid Hydroperoxide Glutathione Peroxidase genetics, Immunotherapy methods, Single-Cell Analysis, Gene Expression Regulation, Neoplastic, Xenograft Model Antitumor Assays, Transcriptome, Ferroptosis genetics, Stomach Neoplasms pathology, Stomach Neoplasms immunology, Stomach Neoplasms genetics, Stomach Neoplasms therapy, Tumor Microenvironment immunology, Disease Progression, Adenocarcinoma secondary, Adenocarcinoma immunology, Adenocarcinoma genetics, Adenocarcinoma therapy, Adenocarcinoma pathology, Liver Neoplasms secondary, Liver Neoplasms immunology, Liver Neoplasms genetics, Liver Neoplasms therapy
- Abstract
Background & Aims: Metastases from gastric adenocarcinoma (GAC) lead to high morbidity and mortality. Developing innovative and effective therapies requires a comprehensive understanding of the tumor and immune biology of advanced GAC. Yet, collecting matched specimens from advanced, treatment-naïve patients with GAC poses a significant challenge, limiting the scope of current research, which has focused predominantly on localized tumors. This gap hinders deeper insight into the metastatic dynamics of GAC., Methods: We performed in-depth single-cell transcriptome and immune profiling on 68 paired, treatment-naïve, primary metastatic tumors to delineate alterations in cancer cells and their tumor microenvironment during metastatic progression. To validate our observations, we conducted comprehensive functional studies both in vitro and in vivo, using cell lines and multiple patient-derived xenograft and novel mouse models of GAC., Results: Liver and peritoneal metastases exhibited distinct properties in cancer cells and dynamics of tumor microenvironment phenotypes, supporting the notion that cancer cells and their local tumor microenvironments co-evolve at metastatic sites. Our study also revealed differential activation of cancer meta-programs across metastases. We observed evasion of cancer cell ferroptosis via GPX4 up-regulation during GAC progression. Conditional depletion of Gpx4 or pharmacologic inhibition of ferroptosis resistance significantly attenuated tumor growth and metastatic progression. In addition, ferroptosis-resensitizing treatments augmented the efficacy of chimeric antigen receptor T-cell therapy., Conclusions: This study represents the largest single-cell dataset of metastatic GACs to date. High-resolution mapping of the molecular and cellular dynamics of GAC metastasis has revealed a rationale for targeting ferroptosis defense in combination with chimeric antigen receptor T-cell therapy as a novel therapeutic strategy with potential immense clinical implications., (Copyright © 2024 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. Designing Rigorous and Efficient Clinical Utility Studies for Early Detection Biomarkers.
- Author
-
Zheng Y, Wagner PD, Singal AG, Hanash SM, Srivastava S, Huang Y, Zhao YQ, Chari ST, Marquez G, Etizioni R, Marsh TL, and Feng Z
- Subjects
- Humans, Biomarkers, Tumor blood, Biomarkers, Tumor analysis, Early Detection of Cancer methods, Research Design, Neoplasms diagnosis
- Abstract
Before implementing a biomarker in routine clinical care, it must demonstrate clinical utility by leading to clinical actions that positively affect patient-relevant outcomes. Randomly controlled early detection utility trials, especially those targeting mortality endpoint, are challenging due to their high costs and prolonged duration. Special design considerations are required to determine the clinical utility of early detection assays. This commentary reports on discussions among the National Cancer Institute's Early Detection Research Network investigators, outlining the recommended process for carrying out single-organ biomarker-driven clinical utility studies. We present the early detection utility studies in the context of phased biomarker development. We describe aspects of the studies related to the features of biomarker tests, the clinical context of endpoints, the performance criteria for later phase evaluation, and study size. We discuss novel adaptive design approaches for improving the efficiency and practicality of clinical utility trials. We recommend using multiple strategies, including adopting real-world evidence, emulated trials, and mathematical modeling to circumvent the challenges in conducting early detection utility trials., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
15. Glycosylation in cancer as a source of biomarkers.
- Author
-
Khorami-Sarvestani S, Hanash SM, Fahrmann JF, León-Letelier RA, and Katayama H
- Subjects
- Humans, Glycosylation, Glycoproteins metabolism, Protein Processing, Post-Translational, Animals, Proteomics methods, Neoplasms metabolism, Neoplasms diagnosis, Biomarkers, Tumor metabolism, Polysaccharides metabolism
- Abstract
Introduction: Glycosylation, the process of glycan synthesis and attachment to target molecules, is a crucial and common post-translational modification (PTM) in mammalian cells. It affects the protein's hydrophilicity, charge, solubility, structure, localization, function, and protection from proteolysis. Aberrant glycosylation in proteins can reveal new detection and therapeutic Glyco-biomarkers, which help to improve accurate early diagnosis and personalized treatment. This review underscores the pivotal role of glycans and glycoproteins as a source of biomarkers in human diseases, particularly cancer., Areas Covered: This review delves into the implications of glycosylation, shedding light on its intricate roles in cancer-related cellular processes influencing biomarkers. It is underpinned by a thorough examination of literature up to June 2024 in PubMed, Scopus, and Google Scholar; concentrating on the terms: (Glycosylation[Title/Abstract]) OR (Glycan[Title/Abstract]) OR (glycoproteomics[Title/Abstract]) OR (Proteoglycans[Title/Abstract]) OR (Glycomarkers[Title/Abstract]) AND (Cancer[Title/Abstract]) AND ((Diagno*[Title/Abstract]) OR (Progno*[Title/Abstract]))., Expert Opinion: Glyco-biomarkers enhance early cancer detection, allow early intervention, and improve patient prognoses. However, the abundance and complex dynamic glycan structure may make their scientific and clinical application difficult. This exploration of glycosylation signatures in cancer biomarkers can provide a detailed view of cancer etiology and instill hope in the potential of glycosylation to revolutionize cancer research.
- Published
- 2024
- Full Text
- View/download PDF
16. Multicancer detection tests: What we know and what we don't know.
- Author
-
Hanash SM and Yu PP
- Subjects
- Humans, Mass Screening methods, Mass Screening standards, Early Detection of Cancer methods, Neoplasms diagnosis
- Published
- 2024
- Full Text
- View/download PDF
17. Protein biomarkers and alternatively methylated cell-free DNA detect early stage pancreatic cancer.
- Author
-
Ben-Ami R, Wang QL, Zhang J, Supplee JG, Fahrmann JF, Lehmann-Werman R, Brais LK, Nowak J, Yuan C, Loftus M, Babic A, Irajizad E, Davidi T, Zick A, Hubert A, Neiman D, Piyanzin S, Gal-Rosenberg O, Horn A, Shemer R, Glaser B, Boos N, Jajoo K, Lee L, Clancy TE, Rubinson DA, Ng K, Chabot JA, Kastrinos F, Kluger M, Aguirre AJ, Jänne PA, Bardeesy N, Stanger B, O'Hara MH, Till J, Maitra A, Carpenter EL, Bullock AJ, Genkinger J, Hanash SM, Paweletz CP, Dor Y, and Wolpin BM
- Subjects
- Humans, CA-19-9 Antigen, Biomarkers, Tumor, Pancreas pathology, DNA Methylation, Cell-Free Nucleic Acids metabolism, Pancreatic Neoplasms diagnosis, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Carcinoma, Pancreatic Ductal diagnosis, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology, Adenocarcinoma diagnosis, Adenocarcinoma genetics, Adenocarcinoma pathology
- Abstract
Objective: Pancreatic ductal adenocarcinoma (PDAC) is commonly diagnosed at an advanced stage. Liquid biopsy approaches may facilitate detection of early stage PDAC when curative treatments can be employed., Design: To assess circulating marker discrimination in training, testing and validation patient cohorts (total n=426 patients), plasma markers were measured among PDAC cases and patients with chronic pancreatitis, colorectal cancer (CRC), and healthy controls. Using CA19-9 as an anchor marker, measurements were made of two protein markers (TIMP1, LRG1) and cell-free DNA (cfDNA) pancreas-specific methylation at 9 loci encompassing 61 CpG sites., Results: Comparative methylome analysis identified nine loci that were differentially methylated in exocrine pancreas DNA. In the training set (n=124 patients), cfDNA methylation markers distinguished PDAC from healthy and CRC controls. In the testing set of 86 early stage PDAC and 86 matched healthy controls, CA19-9 had an area under the receiver operating characteristic curve (AUC) of 0.88 (95% CI 0.83 to 0.94), which was increased by adding TIMP1 (AUC 0.92; 95% CI 0.88 to 0.96; p=0.06), LRG1 (AUC 0.92; 95% CI 0.88 to 0.96; p=0.02) or exocrine pancreas-specific cfDNA methylation markers at nine loci (AUC 0.92; 95% CI 0.88 to 0.96; p=0.02). In the validation set of 40 early stage PDAC and 40 matched healthy controls, a combined panel including CA19-9, TIMP1 and a 9-loci cfDNA methylation panel had greater discrimination (AUC 0.86, 95% CI 0.77 to 0.95) than CA19-9 alone (AUC 0.82; 95% CI 0.72 to 0.92)., Conclusion: A combined panel of circulating markers including proteins and methylated cfDNA increased discrimination compared with CA19-9 alone for early stage PDAC., Competing Interests: Competing interests: ABu has received consulting fees from Exelixis and Geistlich Pharma. KM declares research funding from Celgene and Trovagene. AJA has consulted for Oncorus, Inc., Arrakis Therapeutics, and Merck & Co., Inc, and has research funding from Mirati Therapeutics, Syros, Deerfield, Inc., and Novo Ventures that is unrelated to this work. AM is listed as an inventor on a patent that has been licensed by Johns Hopkins University to Thrive Earlier Detection and serves as a consultant for Tezcat Biosciences. BG, RS and YD have filed patents on cfDNA analysis technology, and received research funding from GRAIL. BMW declares research funding from Celgene, Eli Lilly Novartis and Revolution Medicines, and consulting for Celgene, GRAIL, Ipsen and Mirati., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
18. Baseline Serum Inflammatory Proteins Predict Poor CAR T Outcomes in Diffuse Large B-cell Lymphoma.
- Author
-
Faramand RG, Lee SB, Jain MD, Cao B, Wang X, Rejeski K, Subklewe M, Fahrmann JF, Saini NY, Hanash SM, Kang YP, Chang D, Rodriguez PC, Dean EA, Nishihori T, Shah BD, Lazaryan A, Chavez J, Khimani F, Pinilla-Ibarz JA, Dam M, Reid KM, Corallo SA, Menges M, Hidalgo Vargas M, Mandula JK, Holliday BA, Bachmeier CA, Speth K, Song Q, Mattie M, Locke FL, and Davila ML
- Subjects
- Humans, Adaptor Proteins, Signal Transducing, Antigens, CD19 therapeutic use, Blood Proteins, C-Reactive Protein, Ferritins, Receptors, Chimeric Antigen therapeutic use, Lymphoma, Large B-Cell, Diffuse therapy, Hematologic Neoplasms
- Abstract
A subset of patients with diffuse large B-cell lymphoma (DLBCL) treated with CD19 chimeric antigen receptor (CAR) T-cell therapy have poor clinical outcomes. We report serum proteins associated with severe immune-mediated toxicities and inferior clinical responses in 146 patients with DLBCL treated with axicabtagene ciloleucel. We develop a simple stratification based on pre-lymphodepletion C reactive protein (CRP) and ferritin to classify patients into low-, intermediate-, and high-risk groups. We observe that patients in the high-risk category were more likely to develop grade ≥3 toxicities and had inferior overall and progression-free survival. We sought to validate our findings with two independent international cohorts demonstrating that patients classified as low-risk have excellent efficacy and safety outcomes. Based on routine and readily available laboratory tests that can be obtained prior to lymphodepleting chemotherapy, this simple risk stratification can inform patient selection for CAR T-cell therapy., Significance: CAR T-cell therapy has changed the treatment paradigm for patients with relapsed/refractory hematologic malignancies. Despite encouraging efficacy, a subset of patients have poor clinical outcomes. We show that a simple clinically applicable model using pre-lymphodepletion CRP and ferritin can identify patients at high risk of poor outcomes. This article is featured in Selected Articles from This Issue, p. 80., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
19. Circulating microRNA Panel for Prediction of Recurrence and Survival in Early-Stage Lung Adenocarcinoma.
- Author
-
Tai MC, Bantis LE, Parhy G, Kato T, Tanaka I, Chow CW, Fujimoto J, Behrens C, Hase T, Kawaguchi K, Fahrmann JF, Ostrin EJ, Yokoi K, Chen-Yoshikawa TF, Hasegawa Y, Hanash SM, Wistuba II, and Taguchi A
- Subjects
- Humans, Biomarkers, Tumor genetics, Circulating MicroRNA genetics, Adenocarcinoma of Lung genetics, MicroRNAs, Lung Neoplasms diagnosis, Lung Neoplasms genetics
- Abstract
Early-stage lung adenocarcinoma (LUAD) patients remain at substantial risk for recurrence and disease-related death, highlighting the unmet need of biomarkers for the assessment and identification of those in an early stage who would likely benefit from adjuvant chemotherapy. To identify circulating miRNAs useful for predicting recurrence in early-stage LUAD, we performed miRNA microarray analysis with pools of pretreatment plasma samples from patients with stage I LUAD who developed recurrence or remained recurrence-free during the follow-up period. Subsequent validation in 85 patients with stage I LUAD resulted in the development of a circulating miRNA panel comprising miR-23a-3p, miR-320c, and miR-125b-5p and yielding an area under the curve (AUC) of 0.776 in predicting recurrence. Furthermore, the three-miRNA panel yielded an AUC of 0.804, with a sensitivity of 45.8% at 95% specificity in the independent test set of 57 stage I and II LUAD patients. The miRNA panel score was a significant and independent factor for predicting disease-free survival ( p < 0.001, hazard ratio [HR] = 1.64, 95% confidence interval [CI] = 1.51-4.22) and overall survival ( p = 0.001, HR = 1.51, 95% CI = 1.17-1.94). This circulating miRNA panel is a useful noninvasive tool to stratify early-stage LUAD patients and determine an appropriate treatment plan with maximal efficacy.
- Published
- 2024
- Full Text
- View/download PDF
20. Modulating a prebiotic food source influences inflammation and immune-regulating gut microbes and metabolites: insights from the BE GONE trial.
- Author
-
Zhang X, Irajizad E, Hoffman KL, Fahrmann JF, Li F, Seo YD, Browman GJ, Dennison JB, Vykoukal J, Luna PN, Siu W, Wu R, Murage E, Ajami NJ, McQuade JL, Wargo JA, Long JP, Do KA, Lampe JW, Basen-Engquist KM, Okhuysen PC, Kopetz S, Hanash SM, Petrosino JF, Scheet P, and Daniel CR
- Subjects
- Humans, Proteomics, Obesity microbiology, Inflammation, Prebiotics, Gastrointestinal Microbiome
- Abstract
Background: Accessible prebiotic foods hold strong potential to jointly target gut health and metabolic health in high-risk patients. The BE GONE trial targeted the gut microbiota of obese surveillance patients with a history of colorectal neoplasia through a straightforward bean intervention., Methods: This low-risk, non-invasive dietary intervention trial was conducted at MD Anderson Cancer Center (Houston, TX, USA). Following a 4-week equilibration, patients were randomized to continue their usual diet without beans (control) or to add a daily cup of study beans to their usual diet (intervention) with immediate crossover at 8-weeks. Stool and fasting blood were collected every 4 weeks to assess the primary outcome of intra and inter-individual changes in the gut microbiome and in circulating markers and metabolites within 8 weeks. This study was registered on ClinicalTrials.gov as NCT02843425, recruitment is complete and long-term follow-up continues., Findings: Of the 55 patients randomized by intervention sequence, 87% completed the 16-week trial, demonstrating an increase on-intervention in diversity [n = 48; linear mixed effect and 95% CI for inverse Simpson index: 0.16 (0.02, 0.30); p = 0.02] and shifts in multiple bacteria indicative of prebiotic efficacy, including increased Faecalibacterium, Eubacterium and Bifidobacterium (all p < 0.05). The circulating metabolome showed parallel shifts in nutrient and microbiome-derived metabolites, including increased pipecolic acid and decreased indole (all p < 0.002) that regressed upon returning to the usual diet. No significant changes were observed in circulating lipoproteins within 8 weeks; however, proteomic biomarkers of intestinal and systemic inflammatory response, fibroblast-growth factor-19 increased, and interleukin-10 receptor-α decreased (p = 0.01)., Interpretation: These findings underscore the prebiotic and potential therapeutic role of beans to enhance the gut microbiome and to regulate host markers associated with metabolic obesity and colorectal cancer, while further emphasizing the need for consistent and sustainable dietary adjustments in high-risk patients., Funding: This study was funded by the American Cancer Society., Competing Interests: Declaration of interests Study beans were independently purchased with funds from the Dry Bean Health Research Program, a peer-reviewed incentive award (to CRD) created by the Northarvest Bean Growers Association, Communique Inc. to identify and encourage researchers that apply for NIH-funding to support studies on beans and human health., (Copyright © 2023 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
21. Blood-based Proteomic Signatures Associated With MEN1-related Duodenopancreatic Neuroendocrine Tumor Progression.
- Author
-
Fahrmann JF, Wasylishen AR, Pieterman CRC, Irajizad E, Vykoukal J, Wu R, Dennison JB, Peterson CB, Zhao H, Do KA, Halperin DM, Agarwal SK, Blau JE, Jha S, Rivero JD, Nilubol N, Walter MF, Welch JM, Weinstein LS, Vriens MR, van Leeuwaarde RS, van Treijen MJC, Valk GD, Perrier ND, Hanash SM, and Katayama H
- Subjects
- Animals, Humans, Mice, Disease Progression, Proteomics, Proto-Oncogene Proteins, Multiple Endocrine Neoplasia Type 1 pathology, Neuroendocrine Tumors pathology, Pancreatic Neoplasms pathology
- Abstract
Purpose: Patients with multiple endocrine neoplasia type 1 (MEN1) are predisposed to develop duodenopancreatic neuroendocrine tumors (dpNETs), and metastatic dpNET is the primary cause of disease-related mortality. Presently, there is a paucity of prognostic factors that can reliably identify patients with MEN1-related dpNETS who are at high risk of distant metastasis. In the current study, we aimed to establish novel circulating molecular protein signatures associated with disease progression., Experimental Design: Mass spectrometry-based proteomic profiling was conducted on plasmas procured through an international collaboration between MD Anderson Cancer Center, the National Institutes of Health, and the University Medical Center Utrecht from a cohort of 56 patients with MEN1 [14 with distant metastasis dpNETs (cases) and 42 with either indolent dpNETs or no dpNETs (controls)]. Findings were compared to proteomic profiles generated from serially collected plasmas from a mouse model of Men1-pancreatic neuroendocrine tumors (Men1fl/flPdx1-CreTg) and control mice (Men1fl/fl)., Results: A total of 187 proteins were found to be elevated in MEN1 patients with distant metastasis compared to controls, including 9 proteins previously associated with pancreatic cancer and other neuronal proteins. Analyses of mouse plasmas revealed 196 proteins enriched for transcriptional targets of oncogenic MYCN, YAP1, POU5F1, and SMAD that were associated with disease progression in Men1fl/flPdx1-CreTg mice. Cross-species intersection revealed 19 proteins positively associated with disease progression in both human patients and in Men1fl/flPdx1-CreTg mice., Conclusions: Our integrated analyses identified novel circulating protein markers associated with disease progression in MEN1-related dpNET., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
22. Tumor-Associated and Systemic Autoimmunity in Pre-Clinical Breast Cancer among Post-Menopausal Women.
- Author
-
Parks CG, Wilson LE, Capello M, Deane KD, and Hanash SM
- Subjects
- Humans, Female, Autoimmunity, Cross-Sectional Studies, Postmenopause, Antibodies, Antinuclear, Breast Neoplasms, Autoimmune Diseases
- Abstract
Autoantibodies to tumor-associated antigens (anti-TAA) are potential biomarkers for breast cancer, but their relationship systemic autoimmunity as ascertained though antinuclear antibodies (ANA) is unknown and warrants consideration given the common occurrence of autoimmunity and autoimmune diseases among women. The relationship between anti-TAAs and ANA among women who were later diagnosed with breast cancer and others who remained cancer free in the Women's Health Initiative cohort. The study sample included 145 post-menopausal women with baseline ANA data. A total of 37 ANA-positive women who developed breast cancer (i.e., cases; mean time to diagnosis 6.8 years [SE 3.9]) were matched to a random sample of 36 ANA-negative cases by age and time to diagnosis. An age-matched control sample was selected including 35 ANA-positive and 37 ANA-negative women who did not develop breast cancer (i.e., controls; follow-up time ~13 years [SE 3]). Baseline sera were assessed for Immunoglobulin G (IgG) antibodies, measured by custom microarray for 171 breast and other cancer-associated TAA. We used linear regression to estimate cross-sectional associations of ANA with log-transformed anti-TAA among cases and controls. Most anti-TAA did not vary by ANA status. Two anti-TAA were elevated in ANA-positive compared to ANA-negative cases: anti-PGM3 ( p = 0.004) and anti-TTN ( p = 0.005, especially in cases up to 7 years before diagnosis, p = 0.002). Anti-TAA antibodies were not generally related to ANA, a common marker of systemic autoimmunity. Associations of ANA with particular antigens inducing autoimmunity prior to breast cancer warrant further investigation.
- Published
- 2023
- Full Text
- View/download PDF
23. Evolution of immune and stromal cell states and ecotypes during gastric adenocarcinoma progression.
- Author
-
Wang R, Song S, Qin J, Yoshimura K, Peng F, Chu Y, Li Y, Fan Y, Jin J, Dang M, Dai E, Pei G, Han G, Hao D, Li Y, Chatterjee D, Harada K, Pizzi MP, Scott AW, Tatlonghari G, Yan X, Xu Z, Hu C, Mo S, Shanbhag N, Lu Y, Sewastjanow-Silva M, Fouad Abdelhakeem AA, Peng G, Hanash SM, Calin GA, Yee C, Mazur P, Marsden AN, Futreal A, Wang Z, Cheng X, Ajani JA, and Wang L
- Subjects
- Humans, Ecotype, Stromal Cells pathology, Tumor Microenvironment, Stomach Neoplasms genetics, Stomach Neoplasms metabolism, Adenocarcinoma pathology, Cancer-Associated Fibroblasts pathology, Precancerous Conditions pathology
- Abstract
Understanding tumor microenvironment (TME) reprogramming in gastric adenocarcinoma (GAC) progression may uncover novel therapeutic targets. Here, we performed single-cell profiling of precancerous lesions, localized and metastatic GACs, identifying alterations in TME cell states and compositions as GAC progresses. Abundant IgA
+ plasma cells exist in the premalignant microenvironment, whereas immunosuppressive myeloid and stromal subsets dominate late-stage GACs. We identified six TME ecotypes (EC1-6). EC1 is exclusive to blood, while EC4, EC5, and EC2 are highly enriched in uninvolved tissues, premalignant lesions, and metastases, respectively. EC3 and EC6, two distinct ecotypes in primary GACs, associate with histopathological and genomic characteristics, and survival outcomes. Extensive stromal remodeling occurs in GAC progression. High SDC2 expression in cancer-associated fibroblasts (CAFs) is linked to aggressive phenotypes and poor survival, and SDC2 overexpression in CAFs contributes to tumor growth. Our study provides a high-resolution GAC TME atlas and underscores potential targets for further investigation., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
24. A Nasal Inflammatory Cytokine Signature Is Associated with Early Graft-versus-Host Disease of the Lung after Allogeneic Hematopoietic Cell Transplantation: Proof of Concept.
- Author
-
Ostrin EJ, Rider NL, Alousi AM, Irajizad E, Li L, Peng Q, Kim ST, Bashoura L, Arain MH, Noor LZ, Patel N, Mehta R, Popat UR, Hosing C, Jenq RR, Rondon G, Hanash SM, Paczesny S, Shpall EJ, Champlin RE, Dickey BF, and Sheshadri A
- Subjects
- Humans, Cytokines, Lung metabolism, Inflammation, Bronchiolitis Obliterans diagnosis, Bronchiolitis Obliterans etiology, Bronchiolitis Obliterans metabolism, Lung Transplantation, Graft vs Host Disease diagnosis, Bronchiolitis Obliterans Syndrome, Hematopoietic Stem Cell Transplantation adverse effects
- Abstract
Respiratory inflammation in bronchiolitis obliterans syndrome (BOS) after hematopoietic cell transplantation (HCT) is poorly understood. Clinical criteria for early-stage BOS (stage 0p) often capture HCT recipients without BOS. Measuring respiratory tract inflammation may help identify BOS, particularly early BOS. We conducted a prospective observational study in HCT recipients with new-onset BOS (n = 14), BOS stage 0p (n = 10), and recipients without lung impairment with (n = 3) or without (n = 8) chronic graft-versus-host disease and measured nasal inflammation using nasosorption at enrollment and then every 3 mo for 1 y. We divided BOS stage 0p into impairment that did not return to baseline values (preBOS, n = 6), or transient impairment (n = 4). We tested eluted nasal mucosal lining fluid from nasosorption matrices for inflammatory chemokines and cytokines using multiplex magnetic bead immunoassays. We analyzed between-group differences using the Kruskal-Wallis method, adjusting for multiple comparisons. We found increased nasal inflammation in preBOS and therefore directly compared patients with preBOS to those with transient impairment, as this would be of greatest diagnostic relevance. After adjusting for multiple corrections, we found significant increases in growth factors (FGF2, TGF-α, GM-CSF, VEGF), macrophage activation (CCL4, TNF-α, IL-6), neutrophil activation (CXCL2, IL-8), T cell activation (CD40 ligand, IL-2, IL-12p70, IL-15), type 2 inflammation (eotaxin, IL-4, IL-13), type 17 inflammation (IL-17A), dendritic maturation (FLT3 ligand, IL-7), and counterregulatory molecules (PD-L1, IL-1 receptor antagonist, IL-10) in preBOS patients compared to transient impairment. These differences waned over time. In conclusion, a transient multifaceted nasal inflammatory response is associated with preBOS. Our findings require validation in larger longitudinal cohorts., (Copyright © 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
25. Proteogenomic landscape of gastric adenocarcinoma peritoneal metastases.
- Author
-
Zhao S, Wang R, Song S, Hao D, Han G, Song X, Zhang J, Pizzi MP, Shanbhag N, Futreal A, Badgwell B, Harada K, Calin G, Vykoukal J, Yu CY, Katayama H, Hanash SM, Wang L, and Ajani JA
- Abstract
Advanced gastric adenocarcinoma (GAC) often leads to peritoneal carcinomatosis (PC) and is associated with very poor outcome. Here we report the comprehensive proteogenomic study of ascites derived cells from a prospective GAC cohort (n = 26 patients with peritoneal carcinomatosis, PC). A total of 16,449 proteins were detected from whole cell extracts (TCEs). Unsupervised hierarchical clustering resulted in three distinct groups that reflected extent of enrichment in tumor cells. Integrated analysis revealed enriched biological pathways and notably, some druggable targets (cancer-testis antigens, kinases, and receptors) that could be exploited to develop effective therapies and/or tumor stratifications. Systematic comparison of expression levels of proteins and mRNAs revealed special expression patterns of key therapeutics target notably high mRNA and low protein expression of HAVCR2 (TIM-3), and low mRNA but high protein expression of cancer-testis antigens CTAGE1 and CTNNA2. These results inform strategies to target GAC vulnerabilities., Competing Interests: The authors declare that they have no competing interests., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
26. Epithelial SOX9 drives progression and metastases of gastric adenocarcinoma by promoting immunosuppressive tumour microenvironment.
- Author
-
Fan Y, Li Y, Yao X, Jin J, Scott A, Liu B, Wang S, Huo L, Wang Y, Wang R, Pool Pizzi M, Ma L, Shao S, Sewastjanow-Silva M, Waters R, Chatterjee D, Liu B, Shanbhag N, Peng G, Calin GA, Mazur PK, Hanash SM, Ishizawa J, Hirata Y, Nagano O, Wang Z, Wang L, Xian W, McKeon F, Ajani JA, and Song S
- Subjects
- Humans, Tumor Microenvironment, Immunosuppressive Agents, Immunosuppression Therapy, SOX9 Transcription Factor genetics, Adenocarcinoma genetics, Adenocarcinoma pathology, Stomach Neoplasms genetics, Stomach Neoplasms pathology
- Abstract
Objective: Many cancers engage embryonic genes for rapid growth and evading the immune system. SOX9 has been upregulated in many tumours, yet the role of SOX9 in mediating immunosuppressive tumour microenvironment is unclear. Here, we aim to dissect the role of SOX9-mediated cancer stemness attributes and immunosuppressive microenvironment in advanced gastric adenocarcinoma (GAC) for novel therapeutic discoveries., Methods: Bulk RNAseq/scRNA-seq, patient-derived cells/models and extensive functional studies were used to identify the expression and functions of SOX9 and its target genes in vitro and in vivo. Immune responses were studied in PBMCs or CD45
+ immune cells cocultured with tumour cells with SOX9high or knockout and the KP-Luc2 syngeneic models were used for efficacy of combinations., Results: SOX9 is one of the most upregulated SOX genes in GAC and highly expressed in primary and metastatic tissues and associated with poor prognosis. Depletion of SOX9 in patient-derived GAC cells significantly decreased cancer stemness attributes, tumour formation and metastases and consistently increased CD8+ T cell responses when cocultured with PBMCs/CD45+ cells from GAC patients. RNA sequencing identified the leukaemia inhibitory factor (LIF) as the top secreted molecule regulated by SOX9 in tumour cells and was enriched in malignant ascites and mediated SOX9-induced M2 macrophage repolarisation and inhibited T cell function., Conclusion: Epithelial SOX9 is critical in suppressing CD8+ T cell responses and modified macrophage function in GAC through the paracrine LIF factor. Cotargeting LIF/LIFR and CSF1R has great potential in targeting SOX9-mediated cancer stemness, T cell immunosuppression and metastases suggesting the novel combination therapy against advanced GAC., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2023. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2023
- Full Text
- View/download PDF
27. Deep Learning Models for Automated Assessment of Breast Density Using Multiple Mammographic Image Types.
- Author
-
Rigaud B, Weaver OO, Dennison JB, Awais M, Anderson BM, Chiang TD, Yang WT, Leung JWT, Hanash SM, and Brock KK
- Abstract
Recently, convolutional neural network (CNN) models have been proposed to automate the assessment of breast density, breast cancer detection or risk stratification using single image modality. However, analysis of breast density using multiple mammographic types using clinical data has not been reported in the literature. In this study, we investigate pre-trained EfficientNetB0 deep learning (DL) models for automated assessment of breast density using multiple mammographic types with and without clinical information to improve reliability and versatility of reporting. 120,000 for-processing and for-presentation full-field digital mammograms (FFDM), digital breast tomosynthesis (DBT), and synthesized 2D images from 5032 women were retrospectively analyzed. Each participant underwent up to 3 screening examinations and completed a questionnaire at each screening encounter. Pre-trained EfficientNetB0 DL models with or without clinical history were optimized. The DL models were evaluated using BI-RADS (fatty, scattered fibroglandular densities, heterogeneously dense, or extremely dense) versus binary (non-dense or dense) density classification. Pre-trained EfficientNetB0 model performances were compared using inter-observer and commercial software (Volpara) variabilities. Results show that the average Fleiss' Kappa score between-observers ranged from 0.31-0.50 and 0.55-0.69 for the BI-RADS and binary classifications, respectively, showing higher uncertainty among experts. Volpara-observer agreement was 0.33 and 0.54 for BI-RADS and binary classifications, respectively, showing fair to moderate agreement. However, our proposed pre-trained EfficientNetB0 DL models-observer agreement was 0.61-0.66 and 0.70-0.75 for BI-RADS and binary classifications, respectively, showing moderate to substantial agreement. Overall results show that the best breast density estimation was achieved using for-presentation FFDM and DBT images without added clinical information. Pre-trained EfficientNetB0 model can automatically assess breast density from any images modality type, with the best results obtained from for-presentation FFDM and DBT, which are the most common image archived in clinical practice.
- Published
- 2022
- Full Text
- View/download PDF
28. Contributions of Circulating microRNAs for Early Detection of Lung Cancer.
- Author
-
Vykoukal J, Fahrmann JF, Patel N, Shimizu M, Ostrin EJ, Dennison JB, Ivan C, Goodman GE, Thornquist MD, Barnett MJ, Feng Z, Calin GA, and Hanash SM
- Abstract
There is unmet need to develop circulating biomarkers that would enable earlier interception of lung cancer when more effective treatment options are available. Here, a set of 30 miRNAs, selected from a review of the published literature were assessed for their predictive performance in identifying lung cancer cases in the pre-diagnostic setting. The 30 miRNAs were assayed using sera collected from 102 individuals diagnosed with lung cancer within one year following blood draw and 212 controls matched for age, sex, and smoking status. The additive performance of top-performing miRNA candidates in combination with a previously validated four-protein marker panel (4MP) consisting of the precursor form of surfactant protein B (Pro-SFTPB), cancer antigen 125 (CA125), carcinoembryonic antigen (CEA) and cytokeratin-19 fragment (CYFRA21-1) was additionally assessed. Of the 30 miRNAs evaluated, five (miR-320a-3p, miR-210-3p, miR-92a-3p, miR-21-5p, and miR-140-3p) were statistically significantly (Wilcoxon rank sum test p < 0.05) elevated in case sera compared to controls, with individual AUCs ranging from 0.57−0.62. Compared to the 4MP alone, the combination of 3-miRNAs + 4MP improved sensitivity at 95% specificity by 19.1% ((95% CI of difference 0.0−28.6); two-sided p: 0.006). Our findings demonstrate utility for miRNAs for early detection of lung cancer in combination with a four-protein marker panel.
- Published
- 2022
- Full Text
- View/download PDF
29. A Comprehensive Search of Non-Canonical Proteins in Non-Small Cell Lung Cancer and Their Impact on the Immune Response.
- Author
-
Irajizad E, Fahrmann JF, Long JP, Vykoukal J, Kobayashi M, Capello M, Yu CY, Cai Y, Hsiao FC, Patel N, Park S, Peng Q, Dennison JB, Kato T, Tai MC, Taguchi A, Kadara H, Wistuba II, Katayama H, Do KA, Hanash SM, and Ostrin EJ
- Subjects
- Humans, Immunity, Proteins, Proteomics methods, Adenocarcinoma of Lung pathology, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms
- Abstract
There is substantial interest in mining neoantigens for cancer applications. Non-canonical proteins resulting from frameshift mutations have been identified as neoantigens in cancer. We investigated the landscape of non-canonical proteins in non-small cell lung cancer (NSCLC) and their induced immune response in the form of autoantibodies. A database of cryptoproteins was computationally constructed and comprised all alternate open reading frames (altORFs) and ORFs identified in pseudogenes, noncoding RNAs, and untranslated regions of mRNAs that did not align with known canonical proteins. Proteomic profiles of seventeen lung adenocarcinoma (LUAD) cell lines were searched to evaluate the occurrence of cryptoproteins. To assess the immunogenicity, immunoglobulin (Ig)-bound cryptoproteins in plasmas were profiled by mass spectrometry. The specimen set consisted of plasmas from 30 newly diagnosed NSCLC cases, pre-diagnostic plasmas from 51 NSCLC cases, and 102 control plasmas. An analysis of LUAD cell lines identified 420 cryptoproteins. Plasma Ig-bound analyses revealed 90 cryptoproteins uniquely found in cases and 14 cryptoproteins that had a fold-change >2 compared to controls. In pre-diagnostic samples, 17 Ig-bound cryptoproteins yielded an odds ratio ≥2. Eight Ig-bound cryptoproteins were elevated in both pre-diagnostic and newly diagnosed cases compared to controls. Cryptoproteins represent a class of neoantigens that induce an autoantibody response in NSCLC.
- Published
- 2022
- Full Text
- View/download PDF
30. Blood-Based Biomarker Panel for Personalized Lung Cancer Risk Assessment.
- Author
-
Fahrmann JF, Marsh T, Irajizad E, Patel N, Murage E, Vykoukal J, Dennison JB, Do KA, Ostrin E, Spitz MR, Lam S, Shete S, Meza R, Tammemägi MC, Feng Z, and Hanash SM
- Subjects
- Clinical Trials as Topic, Humans, Lung, Male, Mass Screening methods, Risk Assessment methods, Early Detection of Cancer methods, Lung Neoplasms diagnosis
- Abstract
Purpose: To investigate whether a panel of circulating protein biomarkers would improve risk assessment for lung cancer screening in combination with a risk model on the basis of participant characteristics., Methods: A blinded validation study was performed using prostate lung colorectal ovarian (PLCO) Cancer Screening Trial data and biospecimens to evaluate the performance of a four-marker protein panel (4MP) consisting of the precursor form of surfactant protein B, cancer antigen 125, carcinoembryonic antigen, and cytokeratin-19 fragment in combination with a lung cancer risk prediction model (PLCO
m2012 ) compared with current US Preventive Services Task Force (USPSTF) screening criteria. The 4MP was assayed in 1,299 sera collected preceding lung cancer diagnosis and 8,709 noncase sera., Results: The 4MP alone yielded an area under the receiver operating characteristic curve of 0.79 (95% CI, 0.77 to 0.82) for case sera collected within 1-year preceding diagnosis and 0.74 (95% CI, 0.72 to 0.76) among the entire specimen set. The combined 4MP + PLCOm2012 model yielded an area under the receiver operating characteristic curve of 0.85 (95% CI, 0.82 to 0.88) for case sera collected within 1 year preceding diagnosis. The benefit of the 4MP in the combined model resulted from improvement in sensitivity at high specificity. Compared with the USPSTF2021 criteria, the combined 4MP + PLCOm2012 model exhibited statistically significant improvements in sensitivity and specificity. Among PLCO participants with ≥ 10 smoking pack-years, the 4MP + PLCOm2012 model would have identified for annual screening 9.2% more lung cancer cases and would have reduced referral by 13.7% among noncases compared with USPSTF2021 criteria., Conclusion: A blood-based biomarker panel in combination with PLCOm2012 significantly improves lung cancer risk assessment for lung cancer screening., Competing Interests: Johannes F. FahrmannPatents, Royalties, Other Intellectual Property: There is IP related to biomarkers for early detection of pancreas cancer Nikul PatelPatents, Royalties, Other Intellectual Property: The Lung IP Panel Jennifer B. DennisonResearch Funding: Cosmos Wisdom (Inst), Dynex (Inst)Patents, Royalties, Other Intellectual Property: IP on pancreas cancer early detection biomarkers, IP on lung cancer nodules early detection biomarkers Edwin OstrinHonoraria: AstraZeneca/MedImmuneOpen Payments Link: https://openpaymentsdata.cms.gov/physician/877532 Stephen LamResearch Funding: Nucleix Inc (Inst)Patents, Royalties, Other Intellectual Property: Deep learning prediction algorithm to estimate the 3-year lung cancer risk and cancer-related mortality for individuals who have two or more screening chest CT scans. Joint application by Johns Hopkins University and the BC Cancer Agency Patent pending (Inst) Sanjay SheteStock and Other Ownership Interests: Vertex Martin C. TammemägiConsulting or Advisory Role: AstraZeneca, Nucleix, Medial EarlySign Ziding FengResearch Funding: Exact Sciences (Inst)Patents, Royalties, Other Intellectual Property: I am one of the coinventors for a biomarker panel for pancreatic cancer. The patent was filed by the UT MD Anderson Cancer Center and was licensed to a company by the UT MD Anderson Cancer Center; I am a coinventor for a biomarker test. UT MDACCs own the IP. I received a license fee in January 2019. No other payment has been received after that Samir HanashHonoraria: Abbott Laboratories, BMSResearch Funding: Cosmos Wisdom, DynexPatents, Royalties, Other Intellectual Property: Patents submitted for lung and pancreatic cancer diagnostic markers (Inst)No other potential conflicts of interest were reported.- Published
- 2022
- Full Text
- View/download PDF
31. CES2 sustains HNF4α expression to promote pancreatic adenocarcinoma progression through an epoxide hydrolase-dependent regulatory loop.
- Author
-
Chen Y, Capello M, Rios Perez MV, Vykoukal JV, Roife D, Kang Y, Prakash LR, Katayama H, Irajizad E, Fleury A, Ferri-Borgogno S, Baluya DL, Dennison JB, Do KA, Fiehn O, Maitra A, Wang H, Chiao PJ, Katz MHG, Fleming JB, Hanash SM, and Fahrmann JF
- Subjects
- Animals, Carboxylesterase genetics, Cell Line, Tumor, Epoxide Hydrolases genetics, Epoxide Hydrolases therapeutic use, Humans, Mice, Adenocarcinoma genetics, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism
- Abstract
Objective: Intra-tumoral expression of the serine hydrolase carboxylesterase 2 (CES2) contributes to the activation of the pro-drug irinotecan in pancreatic ductal adenocarcinoma (PDAC). Given other potential roles of CES2, we assessed its regulation, downstream effects, and contribution to tumor development in PDAC., Methods: Association between the mRNA expression of CES2 in pancreatic tumors and overall survival was assessed using The Cancer Genome Atlas. Cell viability, clonogenic, and anchorage-independent growth assays as well as an orthotopic mouse model of PDAC were used to evaluate the biological relevance of CES2 in pancreatic cancer. CES2-driven metabolic changes were determined by untargeted and targeted metabolomic analyses., Results: Elevated tumoral CES2 mRNA expression was a statistically significant predictor of poor overall survival in PDAC patients. Knockdown of CES2 in PDAC cells reduced cell viability, clonogenic capacity, and anchorage-independent growth in vitro and attenuated tumor growth in an orthotopic mouse model of PDAC. Mechanistically, CES2 was found to promote the catabolism of phospholipids resulting in HNF4α activation through a soluble epoxide hydrolase (sEH)-dependent pathway. Targeting of CES2 via siRNA or small molecule inhibitors attenuated HNF4α protein expression and reduced gene expression of classical/progenitor markers and increased basal-like markers. Targeting of the CES2-sEH-HNF4α axis using small molecule inhibitors of CES2 or sEH reduced cell viability., Conclusions: We establish a novel regulatory loop between CES2 and HNF4α to sustain the progenitor subtype and promote PDAC progression and highlight the potential utility of CES2 or sEH inhibitors for the treatment of PDAC as part of non-irinotecan-containing regimens., (Copyright © 2021 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
32. Proteomic Profiling of the Tumor Microenvironment.
- Author
-
Capello M, Katayama H, and Hanash SM
- Subjects
- Extracellular Matrix metabolism, Mass Spectrometry, Proteins metabolism, Proteomics, Tumor Microenvironment
- Abstract
The tumor microenvironment forms a complex pro-tumorigenic milieu constituted by extracellular matrix, surrounding stroma, infiltrating cell populations, and signaling molecules. Proteomic studies have the potential to reveal how individual cell populations within the tumor tissue modulate the microenvironment through protein secretion and consequently alter their protein expression and localization to adapt to this milieu. As a result, proteomic approaches have uncovered how these dynamic components communicate and promote tumor development and progression. The characterization of these mechanisms is relevant for the identification of clinically targetable pathways and for the development of diagnostic tools. Here we describe a method based on the isolation of individual cell compartments and the chromatographic fractionation of intact proteins, followed by enzymatic digestion of individual fractions, and mass-spectrometry analysis, for the profiling of tumor microenvironment cell populations., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
33. A Blood-based Polyamine Signature Associated With MEN1 Duodenopancreatic Neuroendocrine Tumor Progression.
- Author
-
Fahrmann JF, Wasylishen AR, Pieterman CRC, Irajizad E, Vykoukal J, Murage E, Wu R, Dennison JB, Krishna H, Peterson CB, Lozano G, Zhao H, Do KA, Halperin DM, Agarwal SK, Blau JE, Del Rivero J, Nilubol N, Walter MF, Welch JM, Weinstein LS, Vriens MR, van Leeuwaarde RS, van Treijen MJC, Valk GD, Perrier ND, and Hanash SM
- Subjects
- Adult, Aged, Case-Control Studies, Disease Progression, Duodenal Neoplasms blood, Duodenal Neoplasms epidemiology, Female, Follow-Up Studies, Humans, Male, Middle Aged, Multiple Endocrine Neoplasia Type 1 blood, Multiple Endocrine Neoplasia Type 1 epidemiology, Neuroendocrine Tumors blood, Neuroendocrine Tumors epidemiology, Pancreatic Neoplasms blood, Pancreatic Neoplasms epidemiology, Prognosis, Retrospective Studies, United States epidemiology, Young Adult, Biomarkers, Tumor blood, Duodenal Neoplasms pathology, Multiple Endocrine Neoplasia Type 1 pathology, Neuroendocrine Tumors pathology, Pancreatic Neoplasms pathology, Polyamines blood
- Abstract
Context: Duodenopancreatic neuroendocrine tumors (dpNETs) frequently occur in patients with multiple endocrine neoplasia type 1 (MEN1), and metastatic dpNET is the primary cause of disease-related mortality. There is a need for biomarkers that can identify patients with MEN1-related dpNETs that are at high risk of developing distant metastasis. Polyamines have tumor-promoting roles in several cancer types., Objective: We hypothesized that MEN1-dpNET-related disease progression is associated with elevated levels of circulating polyamines., Methods: Through an international collaboration between The University of Texas MD Anderson Cancer Center, the National Institutes of Health, and the University Medical Center Utrecht, plasma polyamine levels were assessed using mass spectrometry in 84 patients with MEN1 (20 with distant metastatic dpNETs [patients] and 64 with either indolent dpNETs or no dpNETs [controls]). A mouse model of MEN1-pNET, Men1fl/flPdx1-CreTg, was used to test time-dependent changes in plasma polyamines associated with disease progression., Results: A 3-marker plasma polyamine signature (3MP: N-acetylputrescine, acetylspermidine, and diacetylspermidine) distinguished patients with metastatic dpNETs from controls in an initial set of plasmas from the 3 participating centers. The fixed 3MP yielded an area under the curve of 0.84 (95% CI, 0.62-1.00) with 66.7% sensitivity at 95% specificity for distinguishing patients from controls in an independent test set from MDACC. In Men1fl/flPdx1-CreTg mice, the 3MP was elevated early and remained high during disease progression., Conclusion: Our findings provide a basis for prospective testing of blood-based polyamines as a potential means for monitoring patients with MEN1 for harboring or developing aggressive disease., (© The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
34. Integrated genomic profiling and modelling for risk stratification in patients with advanced oesophagogastric adenocarcinoma.
- Author
-
Hao D, He S, Harada K, Pizzi MP, Lu Y, Guan P, Chen L, Wang R, Zhang S, Sewastjanow-Silva M, Abdelhakeem A, Shanbhag N, Bhutani M, Han G, Lee JH, Zhao S, Weston B, Blum Murphy M, Waters R, Estrella JS, Roy-Chowdhuri S, Gan Q, Lee JS, Peng G, Hanash SM, Calin GA, Song X, Zhang J, Song S, Wang L, and Ajani JA
- Subjects
- DNA Copy Number Variations, Female, Humans, Male, Prognosis, Risk Assessment, Sequence Analysis, RNA, Exome Sequencing, Adenocarcinoma genetics, Esophageal Neoplasms genetics, Genetic Profile, Stomach Neoplasms genetics
- Abstract
Objective: Prognosis of patients with advanced oesophagogastric adenocarcinoma (mEGAC) is poor and molecular determinants of shorter or longer overall survivors are lacking. Our objective was to identify molecular features and develop a prognostic model by profiling the genomic features of patients with mEGAC with widely varying outcomes., Design: We profiled 40 untreated mEGACs (20 shorter survivors <13 months and 20 longer survivors >36 months) with whole-exome sequencing (WES) and RNA sequencing and performed an integrated analysis of exome, transcriptome, immune profile and pathological phenotypes to identify the molecular determinants, developing an integrated model for prognosis and comparison with The Cancer Genome Atlas (TCGA) cohorts., Results: KMT2C alterations were exclusively observed in shorter survivors together with high level of intratumour heterogeneity and complex clonal architectures, whereas the APOBEC mutational signatures were significantly enriched in longer survivors. Notably, the loss of heterozygosity in chromosome 4 (Chr4) was associated with shorter survival and 'cold' immune phenotype characterised by decreased B, CD8, natural killer cells and interferon-gamma responses. Unsupervised transcriptomic clustering revealed a shorter survivor subtype with distinct expression features (eg, upregulated druggable targets JAK2, MAP3K13 and MECOM ). An integrated model was then built based on clinical variables and the identified molecular determinants, which significantly segregated shorter and longer survivors. All the above features and the integrated model have been validated independently in multiple TCGA cohorts., Conclusion: This study discovered novel molecular features prognosticating overall survival in patients with mEGAC and identified potential novel targets in shorter survivors., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
35. Combinatorial Effect of PLK1 Inhibition with Temozolomide and Radiation in Glioblastoma.
- Author
-
Pandey A, Tripathi SC, Mai J, Hanash SM, Shen H, Mitra S, and Rostomily RC
- Abstract
New strategies that improve median survivals of only ~15-20 months for glioblastoma (GBM) with the current standard of care (SOC) which is concurrent temozolomide (TMZ) and radiation (XRT) treatment are urgently needed. Inhibition of polo-like kinase 1 (PLK1), a multifunctional cell cycle regulator, overexpressed in GBM has shown therapeutic promise but has never been tested in the context of SOC. Therefore, we examined the mechanistic and therapeutic impact of PLK1 specific inhibitor (volasertib) alone and in combination with TMZ and/or XRT on GBM cells. We quantified the effects of volasertib alone and in combination with TMZ and/or XRT on GBM cell cytotoxicity/apoptosis, mitochondrial membrane potential (MtMP), reactive oxygen species (ROS), cell cycle, stemness, DNA damage, DNA repair genes, cellular signaling and in-vivo tumor growth. Volasertib alone and in combination with TMZ and/or XRT promoted apoptotic cell death, altered MtMP, increased ROS and G2/M cell cycle arrest. Combined volasertib and TMZ treatment reduced side population (SP) indicating activity against GBM stem-like cells. Volasertib combinatorial treatment also significantly increased DNA damage and reduced cell survival by inhibition of DNA repair gene expression and modulation of ERK/MAPK, AMPK and glucocorticoid receptor signaling. Finally, as observed in-vitro, combined volasertib and TMZ treatment resulted in synergistic inhibition of tumor growth in-vivo. Together these results identify new mechanisms of action for volasertib that provide a strong rationale for further investigation of PLK1 inhibition as an adjunct to current GBM SOC therapy.
- Published
- 2021
- Full Text
- View/download PDF
36. The Transcriptomic Landscape of Mismatch Repair-Deficient Intestinal Stem Cells.
- Author
-
Bommi PV, Bowen CM, Reyes-Uribe L, Wu W, Katayama H, Rocha P, Parra ER, Francisco-Cruz A, Ozcan Z, Tosti E, Willis JA, Wu H, Taggart MW, Burks JK, Lynch PM, Edelmann W, Scheet PA, Wistuba II, Sinha KM, Hanash SM, and Vilar E
- Subjects
- Animals, Apoptosis, Carcinogenesis genetics, Carcinogenesis metabolism, Cell Proliferation, Colorectal Neoplasms, Hereditary Nonpolyposis genetics, Colorectal Neoplasms, Hereditary Nonpolyposis metabolism, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, MutS Homolog 2 Protein physiology, Prognosis, Proteome analysis, Proteome metabolism, Receptors, G-Protein-Coupled physiology, Stem Cells metabolism, Survival Rate, Tumor Cells, Cultured, Carcinogenesis pathology, Colorectal Neoplasms, Hereditary Nonpolyposis pathology, DNA Mismatch Repair, Gene Expression Regulation, Neoplastic, Intestines physiopathology, Stem Cells pathology, Transcriptome
- Abstract
Lynch syndrome is the most common cause of hereditary colorectal cancer and is secondary to germline alterations in one of four DNA mismatch repair (MMR) genes. Here we aimed to provide novel insights into the initiation of MMR-deficient (MMRd) colorectal carcinogenesis by characterizing the expression profile of MMRd intestinal stem cells (ISC). A tissue-specific MMRd mouse model (Villin-Cre;Msh2
LoxP/LoxP ) was crossed with a reporter mouse ( Lgr5-EGFP-IRES-creERT2 ) to trace and isolate ISCs (Lgr5+) using flow cytometry. Three different ISC genotypes ( Msh2 -KO, Msh2 -HET, and Msh2 -WT) were isolated and processed for mRNA-seq and mass spectrometry, followed by bioinformatic analyses to identify expression signatures of complete MMRd and haplo-insufficiency. These findings were validated using qRT-PCR, IHC, and whole transcriptomic sequencing in mouse tissues, organoids, and a cohort of human samples, including normal colorectal mucosa, premalignant lesions, and early-stage colorectal cancers from patients with Lynch syndrome and patients with familial adenomatous polyposis (FAP) as controls. Msh2 -KO ISCs clustered together with differentiated intestinal epithelial cells from all genotypes. Gene-set enrichment analysis indicated inhibition of replication, cell-cycle progression, and the Wnt pathway and activation of epithelial signaling and immune reaction. An expression signature derived from MMRd ISCs successfully distinguished MMRd neoplastic lesions of patients with Lynch syndrome from FAP controls. SPP1 was specifically upregulated in MMRd ISCs and colocalized with LGR5 in Lynch syndrome colorectal premalignant lesions and tumors. These results show that expression signatures of MMRd ISC recapitulate the initial steps of Lynch syndrome carcinogenesis and have the potential to unveil novel biomarkers of early cancer initiation. SIGNIFICANCE: The transcriptomic and proteomic profile of MMR-deficient intestinal stem cells displays a unique set of genes with potential roles as biomarkers of cancer initiation and early progression., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
37. A Deep Learning Approach to Re-create Raw Full-Field Digital Mammograms for Breast Density and Texture Analysis.
- Author
-
Shu H, Chiang T, Wei P, Do KA, Lesslie MD, Cohen EO, Srinivasan A, Moseley TW, Chang Sen LQ, Leung JWT, Dennison JB, Hanash SM, and Weaver OO
- Abstract
Purpose: To develop a computational approach to re-create rarely stored for-processing (raw) digital mammograms from routinely stored for-presentation (processed) mammograms., Materials and Methods: In this retrospective study, pairs of raw and processed mammograms collected in 884 women (mean age, 57 years ± 10 [standard deviation]; 3713 mammograms) from October 5, 2017, to August 1, 2018, were examined. Mammograms were split 3088 for training and 625 for testing. A deep learning approach based on a U-Net convolutional network and kernel regression was developed to estimate the raw images. The estimated raw images were compared with the originals by four image error and similarity metrics, breast density calculations, and 29 widely used texture features., Results: In the testing dataset, the estimated raw images had small normalized mean absolute error (0.022 ± 0.015), scaled mean absolute error (0.134 ± 0.078) and mean absolute percentage error (0.115 ± 0.059), and a high structural similarity index (0.986 ± 0.007) for the breast portion compared with the original raw images. The estimated and original raw images had a strong correlation in breast density percentage (Pearson r = 0.946) and a strong agreement in breast density grade (Cohen κ = 0.875). The estimated images had satisfactory correlations with the originals in 23 texture features (Pearson r ≥ 0.503 or Spearman ρ ≥ 0.705) and were well complemented by processed images for the other six features., Conclusion: This deep learning approach performed well in re-creating raw mammograms with strong agreement in four image evaluation metrics, breast density, and the majority of 29 widely used texture features. Keywords: Mammography, Breast, Supervised Learning, Convolutional Neural Network (CNN), Deep learning algorithms, Machine Learning AlgorithmsSee also the commentary by Chan in this issue. Supplemental material is available for this article. ©RSNA, 2021., Competing Interests: Disclosures of Conflicts of Interest: H.S. disclosed no relevant relationships. T.C. Activities related to the present article: institution received National Institutes of Health (NIH)/National Cancer Institute (NCI) Cancer Center Support Grant (P30 CA016672); institution supported by Little Green Book Foundation, Center for Global Early Detection at MD Anderson, and McCombs Institute at MD Anderson. Activities not related to the present article: disclosed no relevant relationships. Other relationships: disclosed no relevant relationships. P.W. Activities related to the present article: institution received NIH/NCI Cancer Center Support Grant (P30 CA016672). Activities not related to the present article: disclosed no relevant relationships. K.A.D. disclosed no relevant relationships. M.D.L. disclosed no relevant relationships. E.O.C. disclosed no relevant relationships. A.S. disclosed no relevant relationships. T.W.M. Activities related to the present article: institution received NIH/NCI Cancer Center Support Grant (P30 CA016672). Activities not related to the present article: author is paid medical consultant for Hologic and Merit Medical. Other relationships: disclosed no relevant relationships. L.C.S. Activities related to the present article: institution received NIH/NCI Cancer Center Support Grant (P30 CA016672); institution received support from Little Green Book Foundation, Center for Global Early Detection at MD Anderson, and McCombs Institute at MD Anderson. Activities not related to the present article: disclosed no relevant relationships. Other relationships: disclosed no relevant relationships. J.W.T.L. Activities related to the present article: disclosed no relevant relationships. Activities not related to the present article: author paid by Fujifilm and GE Healthcare for lectures; author has stock/stock options in Subtle Medical (start-up company, stock/stock options have no monetary value at this time). Other relationships: disclosed no relevant relationships. J.B.D. Activities related to the present article: institution supported by Little Green Book Foundation (has supported breast cancer mammography clinical research for the early detection, the MERIT program). Activities not related to the present article: employed by MD Anderson. Other relationships: disclosed no relevant relationships. S.M.H. disclosed no relevant relationships. O.O.W. Activities related to the present article: institution received NIH/NCI Cancer Center Support Grant (P30 CA016672); institution received support from Little Green Book Foundation (sponsor of the patient cohort retrospectively used in the study). Activities not related to the present article: disclosed no relevant relationships. Other relationships: disclosed no relevant relationships., (2021 by the Radiological Society of North America, Inc.)
- Published
- 2021
- Full Text
- View/download PDF
38. Contribution of a Blood-Based Protein Biomarker Panel to the Classification of Indeterminate Pulmonary Nodules.
- Author
-
Ostrin EJ, Bantis LE, Wilson DO, Patel N, Wang R, Kundnani D, Adams-Haduch J, Dennison JB, Fahrmann JF, Chiu HT, Gazdar A, Feng Z, Yuan JM, and Hanash SM
- Subjects
- Biomarkers, Tumor, Case-Control Studies, Humans, Lung Neoplasms diagnosis, Multiple Pulmonary Nodules diagnosis, Solitary Pulmonary Nodule diagnosis
- Abstract
Rationale: The workup and longitudinal monitoring for subjects presenting with pulmonary nodules is a pressing clinical problem. A blood-based biomarker panel potentially has utility for identifying subjects at higher risk for harboring a malignant nodule for whom additional workup would be indicated or subjects at reduced risk for whom imaging-based follow-up would be indicated., Objectives: To assess whether a previously described four-protein biomarker panel, reported to improve assessment of lung cancer risk compared with a smoking-based lung cancer risk model, can provide discrimination between benign and malignant indeterminate pulmonary nodules., Methods: A previously validated multiplex enzyme-linked immunoassay was performed on matched case and control samples from each cohort., Measurements: The biomarker panel was tested in two case-control cohorts of patients presenting with indeterminate pulmonary nodules at the University of Pittsburgh Medical Center and the University of Texas Southwestern., Main Results: In both cohorts, the biomarker panel resulted in improved prediction of lung cancer risk over a model on the basis of nodule size alone. Of particular note, the addition of the marker panel to nodule size greatly improved sensitivity at a high specificity in both cohorts., Conclusions: A four-marker biomarker panel, previously validated to improve lung cancer risk prediction, was found to also have utility in distinguishing benign from malignant indeterminate pulmonary nodules. Its performance in improving sensitivity at a high specificity indicates potential utility of the marker panel in assessing likelihood of malignancy in otherwise indeterminate nodules., (Copyright © 2020 International Association for the Study of Lung Cancer. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
39. YAP1 mediates gastric adenocarcinoma peritoneal metastases that are attenuated by YAP1 inhibition.
- Author
-
Ajani JA, Xu Y, Huo L, Wang R, Li Y, Wang Y, Pizzi MP, Scott A, Harada K, Ma L, Yao X, Jin J, Zhao W, Dong X, Badgwell BD, Shanbhag N, Tatlonghari G, Estrella JS, Roy-Chowdhuri S, Kobayashi M, Vykoukal JV, Hanash SM, Calin GA, Peng G, Lee JS, Johnson RL, Wang Z, Wang L, and Song S
- Subjects
- Animals, Cell Culture Techniques, Humans, Mice, Xenograft Model Antitumor Assays, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing physiology, Adenocarcinoma secondary, Peritoneal Neoplasms secondary, Stomach Neoplasms pathology
- Abstract
Objective: Peritoneal carcinomatosis (PC; malignant ascites or implants) occurs in approximately 45% of advanced gastric adenocarcinoma (GAC) patients and associated with a poor survival. The molecular events leading to PC are unknown. The yes-associated protein 1 ( YAP1 ) oncogene has emerged in many tumour types, but its clinical significance in PC is unclear. Here, we investigated the role of YAP1 in PC and its potential as a therapeutic target., Methods: Patient-derived PC cells, patient-derived xenograft (PDX) and patient-derived orthotopic (PDO) models were used to study the function of YAP1 in vitro and in vivo. Immunofluorescence and immunohistochemical staining, RNA sequencing (RNA-Seq) and single-cell RNA-Seq (sc-RNA-Seq) were used to elucidate the expression of YAP1 and PC cell heterogeneity. LentiCRISPR/Cas9 knockout of YAP1 and a YAP1 inhibitor were used to dissect its role in PC metastases., Results: YAP1 was highly upregulated in PC tumour cells, conferred cancer stem cell (CSC) properties and appeared to be a metastatic driver. Dual staining of YAP1/EpCAM and sc-RNA-Seq revealed that PC tumour cells were highly heterogeneous, YAP1
high PC cells had CSC-like properties and easily formed PDX/PDO tumours but also formed PC in mice, while genetic knockout YAP1 significantly slowed tumour growth and eliminated PC in PDO model. Additionally, pharmacologic inhibition of YAP1 specifically reduced CSC-like properties and suppressed tumour growth in YAP1high PC cells especially in combination with cytotoxics in vivo PDX model., Conclusions: YAP1 is essential for PC that is attenuated by YAP1 inhibition. Our data provide a strong rationale to target YAP1 in clinic for GAC patients with PC., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2021
- Full Text
- View/download PDF
40. Biomarkers and Strategy to Detect Preinvasive and Early Pancreatic Cancer: State of the Field and the Impact of the EDRN.
- Author
-
Liu Y, Kaur S, Huang Y, Fahrmann JF, Rinaudo JA, Hanash SM, Batra SK, Singhi AD, Brand RE, Maitra A, and Haab BB
- Subjects
- Aged, Female, Humans, Male, Pancreatic Neoplasms, Biomarkers, Tumor metabolism, Early Detection of Cancer methods, Pancreatic Neoplasms diagnosis
- Abstract
Patients afflicted with pancreatic ductal adenocarcinoma (PDAC) face a dismal prognosis, but headway could be made if physicians could identify the disease earlier. A compelling strategy to broaden the use of surveillance for PDAC is to incorporate molecular biomarkers in combination with clinical analysis and imaging tools. This article summarizes the components involved in accomplishing biomarker validation and an analysis of the requirements of molecular biomarkers for disease surveillance. We highlight the significance of consortia for this research and highlight resources and infrastructure of the Early Detection Research Network (EDRN). The EDRN brings together the multifaceted expertise and resources needed for biomarker validation, such as study design, clinical care, biospecimen collection and handling, molecular technologies, and biostatistical analysis, and studies coming out of the EDRN have yielded biomarkers that are moving forward in validation. We close the article with an overview of the current investigational biomarkers, an analysis of their performance relative to the established benchmarks, and an outlook on the current needs in the field. The outlook for improving the early detection of PDAC looks promising, and the pace of further research should be quickened through the resources and expertise of the EDRN and other consortia. See all articles in this CEBP Focus section, "NCI Early Detection Research Network: Making Cancer Detection Possible.", (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
41. The Long Noncoding RNA CCAT2 Induces Chromosomal Instability Through BOP1-AURKB Signaling.
- Author
-
Chen B, Dragomir MP, Fabris L, Bayraktar R, Knutsen E, Liu X, Tang C, Li Y, Shimura T, Ivkovic TC, De Los Santos MC, Anfossi S, Shimizu M, Shah MY, Ling H, Shen P, Multani AS, Pardini B, Burks JK, Katayama H, Reineke LC, Huo L, Syed M, Song S, Ferracin M, Oki E, Fromm B, Ivan C, Bhuvaneshwar K, Gusev Y, Mimori K, Menter D, Sen S, Matsuyama T, Uetake H, Vasilescu C, Kopetz S, Parker-Thornburg J, Taguchi A, Hanash SM, Girnita L, Slaby O, Goel A, Varani G, Gagea M, Li C, Ajani JA, and Calin GA
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols pharmacology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Aurora Kinase B metabolism, Azoxymethane toxicity, Carcinogenesis genetics, Cell Line, Tumor, Colon cytology, Colon pathology, Colorectal Neoplasms chemically induced, Colorectal Neoplasms pathology, Cytogenetic Analysis, Dextrans toxicity, Drug Resistance, Neoplasm genetics, Female, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Humans, Intestinal Mucosa cytology, Intestinal Mucosa pathology, Male, Mice, Mice, Transgenic, Neoplasms, Experimental chemically induced, Neoplasms, Experimental pathology, Organoids, Primary Cell Culture, Proto-Oncogene Proteins c-myc metabolism, RNA, Long Noncoding genetics, RNA-Binding Proteins metabolism, Signal Transduction genetics, Chromosomal Instability, Colorectal Neoplasms genetics, Neoplasms, Experimental genetics, RNA, Long Noncoding metabolism, RNA-Binding Proteins genetics
- Abstract
Background & Aims: Chromosomal instability (CIN) is a carcinogenesis event that promotes metastasis and resistance to therapy by unclear mechanisms. Expression of the colon cancer-associated transcript 2 gene (CCAT2), which encodes a long noncoding RNA (lncRNA), associates with CIN, but little is known about how CCAT2 lncRNA regulates this cancer enabling characteristic., Methods: We performed cytogenetic analysis of colorectal cancer (CRC) cell lines (HCT116, KM12C/SM, and HT29) overexpressing CCAT2 and colon organoids from C57BL/6N mice with the CCAT2 transgene and without (controls). CRC cells were also analyzed by immunofluorescence microscopy, γ-H2AX, and senescence assays. CCAT2 transgene and control mice were given azoxymethane and dextran sulfate sodium to induce colon tumors. We performed gene expression array and mass spectrometry to detect downstream targets of CCAT2 lncRNA. We characterized interactions between CCAT2 with downstream proteins using MS2 pull-down, RNA immunoprecipitation, and selective 2'-hydroxyl acylation analyzed by primer extension analyses. Downstream proteins were overexpressed in CRC cells and analyzed for CIN. Gene expression levels were measured in CRC and non-tumor tissues from 5 cohorts, comprising more than 900 patients., Results: High expression of CCAT2 induced CIN in CRC cell lines and increased resistance to 5-fluorouracil and oxaliplatin. Mice that expressed the CCAT2 transgene developed chromosome abnormalities, and colon organoids derived from crypt cells of these mice had a higher percentage of chromosome abnormalities compared with organoids from control mice. The transgenic mice given azoxymethane and dextran sulfate sodium developed more and larger colon polyps than control mice given these agents. Microarray analysis and mass spectrometry indicated that expression of CCAT2 increased expression of genes involved in ribosome biogenesis and protein synthesis. CCAT2 lncRNA interacted directly with and stabilized BOP1 ribosomal biogenesis factor (BOP1). CCAT2 also increased expression of MYC, which activated expression of BOP1. Overexpression of BOP1 in CRC cell lines resulted in chromosomal missegregation errors, and increased colony formation, and invasiveness, whereas BOP1 knockdown reduced viability. BOP1 promoted CIN by increasing the active form of aurora kinase B, which regulates chromosomal segregation. BOP1 was overexpressed in polyp tissues from CCAT2 transgenic mice compared with healthy tissue. CCAT2 lncRNA and BOP1 mRNA or protein were all increased in microsatellite stable tumors (characterized by CIN), but not in tumors with microsatellite instability compared with nontumor tissues. Increased levels of CCAT2 lncRNA and BOP1 mRNA correlated with each other and with shorter survival times of patients., Conclusions: We found that overexpression of CCAT2 in colon cells promotes CIN and carcinogenesis by stabilizing and inducing expression of BOP1 an activator of aurora kinase B. Strategies to target this pathway might be developed for treatment of patients with microsatellite stable colorectal tumors., (Copyright © 2020 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
42. Biomarkers for Lung Cancer Screening and Detection.
- Author
-
Ostrin EJ, Sidransky D, Spira A, and Hanash SM
- Subjects
- Humans, Lung Neoplasms pathology, Biomarkers, Tumor metabolism, Early Detection of Cancer methods, Lung Neoplasms diagnosis
- Abstract
Lung cancer is the leading worldwide cause of cancer mortality, as it is often detected at an advanced stage. Since 2011, low-dose CT scan-based screening has promised a 20% reduction in lung cancer mortality. However, effectiveness of screening has been limited by eligibility only for a high-risk population of heavy smokers and a large number of false positives generated by CT. Biomarkers have tremendous potential to improve early detection of lung cancer by refining lung cancer risk, stratifying positive CT scans, and categorizing intermediate-risk pulmonary nodules. Three biomarker tests (Early CDT-Lung, Nodify XL2, Percepta) have undergone extensive validation and are available to the clinician. The authors discuss these tests, with their clinical applicability and limitations, current ongoing evaluation, and future directions for biomarkers in lung cancer screening and detection. See all articles in this CEBP Focus section, "NCI Early Detection Research Network: Making Cancer Detection Possible.", (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
43. Novel urinary protein biomarker panel for early diagnosis of gastric cancer.
- Author
-
Shimura T, Dayde D, Wang H, Okuda Y, Iwasaki H, Ebi M, Kitagawa M, Yamada T, Yamada T, Hanash SM, Taguchi A, and Kataoka H
- Subjects
- Adult, Aged, Aged, 80 and over, Case-Control Studies, Female, Humans, Male, Middle Aged, Sensitivity and Specificity, Biomarkers, Tumor urine, Early Detection of Cancer methods, Stomach Neoplasms diagnosis, Stomach Neoplasms urine
- Abstract
Background: With the goal of discovering non-invasive biomarkers for early diagnosis of GC, we conducted a case-control study utilising urine samples from individuals with predominantly early GC vs. healthy control (HC)., Methods: Among urine samples from 372 patients, age- and sex-matched 282 patients were randomly divided into three groups: 18 patients in a discovery cohort; 176 patients in a training cohort and 88 patients in a validation cohort., Results: Among urinary proteins identified in the comprehensive quantitative proteomics analysis, urinary levels of TFF1 (uTFF1) and ADAM12 (uADAM12) were significantly independent diagnostic biomarkers for GC, in addition to Helicobacter pylori status. A urinary biomarker panel combining uTFF1, uADAM12 and H. pylori significantly distinguished between HC and GC patients in both training and validation cohorts. On the analysis for sex-specific biomarkers, this combination panel demonstrated a good AUC of 0.858 for male GC, whereas another combination panel of uTFF1, uBARD1 and H. pylori also provided a good AUC of 0.893 for female GC. Notably, each panel could distinguish even stage I GC patients from HC patients (AUC = 0.850 for males; AUC = 0.845 for females)., Conclusions: Novel urinary protein biomarker panels represent promising non-invasive biomarkers for GC, including early-stage disease.
- Published
- 2020
- Full Text
- View/download PDF
44. CES2 Expression in Pancreatic Adenocarcinoma Is Predictive of Response to Irinotecan and Is Associated With Type 2 Diabetes.
- Author
-
Capello M, Fahrmann JF, Rios Perez MV, Vykoukal JV, Irajizad E, Tripathi SC, Roife D, Bantis LE, Kang Y, Kundnani DL, Xu H, Prakash LR, Long JP, Katayama H, Fleury A, Ferri-Borgogno S, Baluya DL, Dennison JB, Aguilar-Bonavides C, Casabar JP, Celiktas M, Do KA, Fiehn O, Maitra A, Wang H, Feng Z, Chiao PJ, Katz MH, Fleming JB, and Hanash SM
- Abstract
Purpose: The combination chemotherapy of fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) has provided clinically meaningful improvement for pancreatic ductal adenocarcinoma (PDAC). We previously uncovered a role for the serine hydrolase carboxylesterase 2 (CES2) in mediating intratumoral activation of the prodrug irinotecan, a constituent of FOLFIRINOX. We aimed to further test the predictive value of CES2 for response to irinotecan using patient-derived xenograft (PDX) models and to elucidate the determinants of CES2 expression and response to FOLFIRINOX treatment among patients with PDAC., Methods: PDXs were engrafted subcutaneously into nude mice and treated for 4 weeks with either saline control or irinotecan. CES2 and hepatocyte nuclear factor 4 alpha (HNF4A) expression in PDAC tissues was evaluated by immunohistochemical and Western blot analysis. Kaplan-Meier and Cox regression analyses were applied to assess the association between overall survival and hemoglobin A1C (HbA1C) levels in patients who underwent neoadjuvant FOLFIRINOX treatment., Results: High CES2 activity in PDAC PDXs was associated with increased sensitivity to irinotecan. Integrated gene expression, proteomic analyses, and in vitro genetic experiments revealed that nuclear receptor HNF4A, which is upregulated in diabetes, is the upstream transcriptional regulator of CES2 expression. Elevated CES2 protein expression in PDAC tissues was positively associated with a history of type 2 diabetes (odds ratio, 4.84; P = .02). High HbA1C levels were associated with longer overall survival in patients who received neoadjuvant FOLFIRINOX treatment ( P = .04)., Conclusion: To our knowledge, we provide, for the first time, evidence that CES2 expression is associated with a history of type 2 diabetes in PDAC and that elevated HbA1C, by predicting tumor CES2 expression, may represent a novel marker for stratifying patients most likely to respond to FOLFIRINOX therapy.
- Published
- 2020
- Full Text
- View/download PDF
45. Therapeutic potential of FLANC, a novel primate-specific long non-coding RNA in colorectal cancer.
- Author
-
Pichler M, Rodriguez-Aguayo C, Nam SY, Dragomir MP, Bayraktar R, Anfossi S, Knutsen E, Ivan C, Fuentes-Mattei E, Lee SK, Ling H, Catela Ivkovic T, Huang G, Huang L, Okugawa Y, Katayama H, Taguchi A, Bayraktar E, Bhattacharya R, Amero P, He WR, Tran AM, Vychytilova-Faltejskova P, Klec C, Bonilla DL, Zhang X, Kapitanovic S, Loncar B, Gafà R, Wang Z, Cristini V, Hanash SM, Bar-Eli M, Lanza G, Slaby O, Goel A, Rigoutsos I, Lopez-Berestein G, and Calin GA
- Subjects
- Animals, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Drug Discovery, Gene Expression Regulation, Neoplastic, Genetic Markers, Genetic Therapy, Humans, Mice, Pharmacogenomic Testing, Vascular Endothelial Growth Factor A metabolism, Carcinogenesis drug effects, Carcinogenesis genetics, Cell Proliferation drug effects, Cell Proliferation genetics, Colorectal Neoplasms genetics, Colorectal Neoplasms therapy, Neovascularization, Pathologic genetics, Neovascularization, Pathologic metabolism, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism, STAT3 Transcription Factor metabolism
- Abstract
Objective: To investigate the function of a novel primate-specific long non-coding RNA (lncRNA), named FLANC, based on its genomic location (co-localised with a pyknon motif), and to characterise its potential as a biomarker and therapeutic target., Design: FLANC expression was analysed in 349 tumours from four cohorts and correlated to clinical data. In a series of multiple in vitro and in vivo models and molecular analyses, we characterised the fundamental biological roles of this lncRNA. We further explored the therapeutic potential of targeting FLANC in a mouse model of colorectal cancer (CRC) metastases., Results: FLANC, a primate-specific lncRNA feebly expressed in normal colon cells, was significantly upregulated in cancer cells compared with normal colon samples in two independent cohorts. High levels of FLANC were associated with poor survival in two additional independent CRC patient cohorts. Both in vitro and in vivo experiments demonstrated that the modulation of FLANC expression influenced cellular growth, apoptosis, migration, angiogenesis and metastases formation ability of CRC cells. In vivo pharmacological targeting of FLANC by administration of 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine nanoparticles loaded with a specific small interfering RNA, induced significant decrease in metastases, without evident tissue toxicity or pro-inflammatory effects. Mechanistically, FLANC upregulated and prolonged the half-life of phosphorylated STAT3, inducing the overexpression of VEGFA, a key regulator of angiogenesis., Conclusions: Based on our findings, we discovered, FLANC as a novel primate-specific lncRNA that is highly upregulated in CRC cells and regulates metastases formation. Targeting primate-specific transcripts such as FLANC may represent a novel and low toxic therapeutic strategy for the treatment of patients., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2020. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
46. NFATc Acts as a Non-Canonical Phenotypic Stability Factor for a Hybrid Epithelial/Mesenchymal Phenotype.
- Author
-
Subbalakshmi AR, Kundnani D, Biswas K, Ghosh A, Hanash SM, Tripathi SC, and Jolly MK
- Abstract
Metastasis remains the cause of over 90% of cancer-related deaths. Cells undergoing metastasis use phenotypic plasticity to adapt to their changing environmental conditions and avoid therapy and immune response. Reversible transitions between epithelial and mesenchymal phenotypes - epithelial-mesenchymal transition (EMT) and its reverse mesenchymal-epithelial transition (MET) - form a key axis of phenotypic plasticity during metastasis and therapy resistance. Recent studies have shown that the cells undergoing EMT/MET can attain one or more hybrid epithelial/mesenchymal (E/M) phenotypes, the process of which is termed as partial EMT/MET. Cells in hybrid E/M phenotype(s) can be more aggressive than those in either epithelial or mesenchymal state. Thus, it is crucial to identify the factors and regulatory networks enabling such hybrid E/M phenotypes. Here, employing an integrated computational-experimental approach, we show that the transcription factor nuclear factor of activated T-cell (NFATc) can inhibit the process of complete EMT, thus stabilizing the hybrid E/M phenotype. It increases the range of parameters enabling the existence of a hybrid E/M phenotype, thus behaving as a phenotypic stability factor (PSF). However, unlike previously identified PSFs, it does not increase the mean residence time of the cells in hybrid E/M phenotypes, as shown by stochastic simulations; rather it enables the co-existence of epithelial, mesenchymal and hybrid E/M phenotypes and transitions among them. Clinical data suggests the effect of NFATc on patient survival in a tissue-specific or context-dependent manner. Together, our results indicate that NFATc behaves as a non-canonical PSF for a hybrid E/M phenotype., (Copyright © 2020 Subbalakshmi, Kundnani, Biswas, Ghosh, Hanash, Tripathi and Jolly.)
- Published
- 2020
- Full Text
- View/download PDF
47. Extracellular Vesicles Mediate B Cell Immune Response and Are a Potential Target for Cancer Therapy.
- Author
-
Kato T, Fahrmann JF, Hanash SM, and Vykoukal J
- Subjects
- Humans, Antigens, Neoplasm immunology, B-Lymphocytes immunology, Extracellular Vesicles immunology, Neoplasms therapy
- Abstract
Extracellular vesicles (EVs) are increasingly understood to participate directly in many essential aspects of host antitumor immune response. Tumor- and immune-cell-derived EVs function in local and systemic contexts with roles in immune processes including cancer antigen conveyance, immune cell priming and activation, as well as immune escape. Current practice of cancer immunotherapy has de facto focused on eliciting T-cell-mediated cytotoxic responses. Humoral immunity is also known to exert antitumor effects, and B cells have been demonstrated to have functions that extend beyond antibody production to include antigen presentation and activation and modulation of T cells and innate immune effectors. Evidence of B cell response against tumor-associated antigens (TAAs) is observed in early stages of tumorigenesis and in most solid tumor types. It is known that EVs convey diverse TAAs, express antigenic-peptide-loaded MHCs, and complex with circulating plasma antitumoral autoantibodies. In this review, we will consider the relationships between EVs, B cells, and other antigen-presenting cells, especially in relation to TAAs. Understanding the intersection of EVs and the cancer immunome will enable opportunities for developing tumor antigen targets, antitumor vaccines and harnessing the full potential of multiple immune system components for next-generation cancer immunotherapies.
- Published
- 2020
- Full Text
- View/download PDF
48. Plasma-Derived Extracellular Vesicles Convey Protein Signatures that Reflect Pathophysiology in Lung and Pancreatic Adenocarcinomas.
- Author
-
Fahrmann JF, Mao X, Irajizad E, Katayama H, Capello M, Tanaka I, Kato T, Wistuba II, Maitra A, Ostrin EJ, Hanash SM, and Vykoukal J
- Abstract
Using a combination of mass-spectrometry and aptamer array-based proteomics, we characterized the protein features of circulating extracellular vesicles (EVs) in the context of lung (LUAD) and pancreatic ductal (PDAC) adenocarcinomas. We profiled EVs isolated from conditioned media of LUAD and PDAC cell lines to identify EV-associated protein cargoes released by these cancer cell types. Analysis of the resulting data identified LUAD and PDAC specific and pan-adenocarcinoma EV protein signatures. Bioinformatic analyses confirmed enrichment of proteins annotated to vesicle-associated processes and intracellular compartments, as well as representation of cancer hallmark functions and processes. Analysis of upstream regulator networks indicated significant enrichment of TP53, MYC, TGFB1 and KRAS-driven network effectors ( p = 1.69 × 10
-77 -2.93 × 10-49 ) manifest in the adenocarcinoma sEV protein cargoes. We extended these findings by profiling the proteome of EVs isolated from lung ( N = 15) and pancreatic ductal ( N = 6) adenocarcinoma patient plasmas obtained at time of diagnosis, along with EVs derived from matched healthy controls ( N = 21). Exploration of these proteomic data revealed abundant protein features in the plasma EVs with capacity to distinguish LUAD and PDAC cases from controls, including features yielding higher performance in the plasma EV isolates relative to unfractionated plasmas.- Published
- 2020
- Full Text
- View/download PDF
49. Immune-Complexome Analysis Identifies Immunoglobulin-Bound Biomarkers That Predict the Response to Chemotherapy of Pancreatic Cancer Patients.
- Author
-
Mandili G, Follia L, Ferrero G, Katayama H, Hong W, Momin AA, Capello M, Giordano D, Spadi R, Satolli MA, Evangelista A, Hanash SM, Cordero F, and Novelli F
- Abstract
Pancreatic Ductal Adenocarcinoma (PDA) is an aggressive malignancy with a very poor outcome. Although chemotherapy (CT) treatment has poor efficacy, it can enhance tumor immunogenicity. Tumor-Associated Antigens (TAA) are self-proteins that are overexpressed in tumors that may induce antibody production and can be PDA theranostic targets. However, the prognostic value of TAA-antibody association as Circulating Immune Complexes (CIC) has not yet been elucidated, mainly due to the lack of techniques that lead to their identification. In this study, we show a novel method to separate IgG, IgM, and IgA CIC from sera to use them as prognostic biomarkers of CT response. The PDA Immune-Complexome (IC) was identified using a LTQ-Orbitrap mass spectrometer followed by computational analysis. The analysis of the IC of 37 PDA patients before and after CT revealed differential associated antigens (DAA) for each immunoglobulin class. Our method identified different PDA-specific CIC in patients that were associated with poor prognosis patients. Finally, CIC levels were significantly modified by CT suggesting that they can be used as effective prognostic biomarkers to follow CT response in PDA patients.
- Published
- 2020
- Full Text
- View/download PDF
50. A Promising CPS1 Inhibitor Keeping Ammonia from Fueling Cancer.
- Author
-
Taguchi A, Fahrmann JF, and Hanash SM
- Subjects
- Carbamoyl-Phosphate Synthase (Ammonia), Humans, Ammonia, Neoplasms drug therapy
- Abstract
Carbamoyl phosphate synthetase 1 (CPS1) drives ammonia conversion to carbamoyl phosphate, and its overexpression supports pyrimidine synthesis and tumor growth, highlighting the potential of CPS1 inhibition as a therapeutic target. In this issue of Cell Chemical Biology, Yao et al. (2020) introduce H3B-120 as a promising novel inhibitor of CPS1., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.