1
|
Cheng TYD, Fu DA, Falzarano SM, Zhang R, Datta S, Zhang W, Omilian AR, Aduse-Poku L, Bian J, Irianto J, Asirvatham JR, Campbell-Thompson M. Association of Computed Tomography Scan-Assessed Body Composition with Immune and PI3K/AKT Pathway Proteins in Distinct Breast Cancer Tumor Components. Int J Mol Sci 2024; 25:13428. [PMID: 39769193 PMCID: PMC11676426 DOI: 10.3390/ijms252413428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
This hypothesis-generating study aims to examine the extent to which computed tomography-assessed body composition phenotypes are associated with immune and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathways in breast tumors. A total of 52 patients with newly diagnosed breast cancer were classified into four body composition types: adequate (lowest two tertiles of total adipose tissue [TAT]) and highest two tertiles of total skeletal muscle [TSM] areas); high adiposity (highest tertile of TAT and highest two tertiles of TSM); low muscle (lowest tertile of TSM and lowest two tertiles of TAT); and high adiposity with low muscle (highest tertile of TAT and lowest tertile of TSM). Immune and PI3K/AKT pathway proteins were profiled in tumor epithelium and the leukocyte-enriched stromal microenvironment using GeoMx (NanoString). Linear mixed models were used to compare log2-transformed protein levels. Compared with the normal type, the low muscle type was associated with higher expression of INPP4B (log2-fold change = 1.14, p = 0.0003, false discovery rate = 0.028). Other significant associations included low muscle type with increased CTLA4 and decreased pan-AKT expression in tumor epithelium, and high adiposity with increased CD3, CD8, CD20, and CD45RO expression in stroma (p < 0.05; false discovery rate > 0.2). With confirmation, body composition can be associated with signaling pathways in distinct components of breast tumors, highlighting the potential utility of body composition in informing tumor biology and therapy efficacies.
Collapse
Affiliation(s)
- Ting-Yuan David Cheng
- Division of Cancer Prevention and Control, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43201, USA
| | - Dongtao Ann Fu
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32611, USA; (D.A.F.); (S.M.F.); (W.Z.); (M.C.-T.)
| | - Sara M. Falzarano
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32611, USA; (D.A.F.); (S.M.F.); (W.Z.); (M.C.-T.)
| | - Runzhi Zhang
- Department of Biostatistics, College of Public Health & Health Professions & College of Medicine, University of Florida, Gainesville, FL 32611, USA; (R.Z.); (S.D.)
| | - Susmita Datta
- Department of Biostatistics, College of Public Health & Health Professions & College of Medicine, University of Florida, Gainesville, FL 32611, USA; (R.Z.); (S.D.)
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32611, USA; (D.A.F.); (S.M.F.); (W.Z.); (M.C.-T.)
| | - Angela R. Omilian
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA;
| | - Livingstone Aduse-Poku
- Department of Epidemiology, College of Public Health & Health Professions & College of Medicine, University of Florida, Gainesville, FL 32611, USA;
| | - Jiang Bian
- Department of Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32611, USA;
| | - Jerome Irianto
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA;
| | | | - Martha Campbell-Thompson
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32611, USA; (D.A.F.); (S.M.F.); (W.Z.); (M.C.-T.)
| |
Collapse
|
2
|
Proskuriakova E, Aryal BB, Shrestha DB, Valencia S, Kovalenko I, Adams M, Boxwala M, Verda L, Khosla PG. Impact of Obesity on Breast Cancer Clinicopathological Characteristics in Underserved US Community Safety-Net Hospital: A Retrospective Single-Center Study. Clin Breast Cancer 2024; 24:e714-e722. [PMID: 39237436 DOI: 10.1016/j.clbc.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/04/2024] [Accepted: 08/07/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Breast cancer continues to pose a significant public health challenge, with its incidence and disproportionate impact on underserved populations in the United States. The relationship between obesity and clinicopathological characteristics at presentation remains a critical area of investigation. Safety-net hospitals caring for underserved communities provide a unique setting to explore these associations. This study seeks to explore a critical gap in knowledge on obesity and breast cancer characteristics in underserved populations in the United States. MATERIALS AND METHODS In this retrospective study, 927 breast cancer patients were included. Analysis was conducted to assess the association between body mass index (BMI), age of diagnosis, tumor clinicopathologic characteristics, and molecular types stratified by menopausal status at diagnosis. Analysis was performed using the Statistical Package for Social Sciences version 29. RESULTS A significant association was found between BMI and menopausal status (P < .05). Disease stage at presentation was significantly associated with BMI (P < .05). Further investigation into BMI categories and tumor characteristics revealed a significant correlation in postmenopausal women, with obesity linked to tumor size and lymph node status (P < .05). No significant associations were observed between HER-2 status, ER/PR status, and obesity in either premenopausal or postmenopausal groups. CONCLUSION This observational retrospective hypothesis-generating study revealed the association between obesity and disease stage and menopause status at diagnosis. In postmenopausal patients, obesity correlated with larger tumor size and advanced lymph node disease involvement. Additionally, ethnic variations were observed, with a higher prevalence of obesity among African American patients.
Collapse
Affiliation(s)
| | - Barun Babu Aryal
- Department of Internal Medicine, Mount Sinai Hospital, Chicago, IL
| | | | | | - Iuliia Kovalenko
- Department of Internal Medicine, UPMC Harrisburg, Harrisburg, PA
| | - Megan Adams
- Department of Internal Medicine, Ross University School of Medicine, Michael, Barbados
| | - Mubaraka Boxwala
- Department of Internal Medicine, Ross University School of Medicine, Michael, Barbados
| | - Larissa Verda
- Department of Internal Medicine, Mount Sinai Hospital, Chicago, IL
| | | |
Collapse
|
3
|
Cheng TYD, Fu DA, Falzarano SM, Zhang R, Datta S, Zhang W, Omilian AR, Aduse-Poku L, Bian J, Irianto J, Asirvatham JR, Campbell-Thompson M. Association of computed tomography scan-assessed body composition with immune and PI3K/AKT pathway proteins in distinct breast cancer tumor components. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.21.24307688. [PMID: 38826360 PMCID: PMC11142286 DOI: 10.1101/2024.05.21.24307688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
This hypothesis-generating study aims to examine the extent to which computed tomography-assessed body composition phenotypes are associated with immune and PI3K/AKT signaling pathways in breast tumors. A total of 52 patients with newly diagnosed breast cancer were classified into four body composition types: adequate (lowest two tertiles of total adipose tissue [TAT]) and highest two tertiles of total skeletal muscle [TSM] areas); high adiposity (highest tertile of TAT and highest two tertiles of TSM); low muscle (lowest tertile of TSM and lowest two tertiles of TAT); and high adiposity with low muscle (highest tertile of TAT and lowest tertile of TSM). Immune and PI3K/AKT pathway proteins were profiled in tumor epithelium and the leukocyte-enriched stromal microenvironment using GeoMx (NanoString). Linear mixed models were used to compare log2-transformed protein levels. Compared with the normal type, the low muscle type was associated with higher expression of INPP4B (log2-fold change = 1.14, p = 0.0003, false discovery rate = 0.028). Other significant associations included low muscle type with increased CTLA4 and decreased pan-AKT expression in tumor epithelium, and high adiposity with increased CD3, CD8, CD20, and CD45RO expression in stroma (P<0.05; false discovery rate >0.2). With confirmation, body composition can be associated with signaling pathways in distinct components of breast tumors, highlighting the potential utility of body composition in informing tumor biology and therapy efficacies.
Collapse
|
4
|
Xiao Y, Liu Y, Sun Y, Huang C, Zhong S. MEIS2 suppresses breast cancer development by downregulating IL10. Cancer Rep (Hoboken) 2024; 7:e2064. [PMID: 38711262 PMCID: PMC11074520 DOI: 10.1002/cnr2.2064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/06/2024] [Accepted: 03/23/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is the most commonly diagnosed female cancer. Homeobox protein MEIS2, a key transcription factor, is involved in the regulation of many developmental and cellular processes. However, the role of MEIS2 in the development of breast cancer is still unclear. AIMS We aimed to examine the role of myeloid ecotropic insertion site (MEIS2) in breast cancer and the association of MEIS2 with breast cancer clinical stages and pathological grades. We revealed the underlying mechanism by which MEIS2 affected breast cancer cell growth and tumor development. METHODS AND RESULTS Using human BC cell lines, clinical samples and animal xenograft model, we reveal that MEIS2 functions as a tumor suppressor in breast cancer. The expression of MEIS2 is inversely correlated with BC clinical stages and pathological grades. MEIS2 knockdown (MEIS2-KD) promotes while MEIS2 overexpression suppresses breast cancer cell proliferation and tumor development in vitro and in animal xenograft models, respectively. To determine the biological function of MEIS2, we screen the expression of a group of MEIS2 potential targeting genes in stable-established cell lines. Results show that the knockdown of MEIS2 in breast cancer cells up-regulates the IL10 expression, but MEIS2 overexpression opposed the effect on IL10 expression. Furthermore, the suppressive role of MEIS2 in breast cancer cell proliferation is associated with the IL10 expression and myeloid cells infiltration. CONCLUSION Our study demonstrates that the tumor suppressor of MEIS2 in breast cancer progression is partially via down regulating the expression of IL10 and promoting myeloid cells infiltration. Targeting MEIS2 would be a potentially therapeutic avenue for BC.
Collapse
Affiliation(s)
- Yongzhi Xiao
- Department of Ultrasound Diagnosis, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yingzhe Liu
- Xiangya International Medical Center, National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yangqing Sun
- Department of Oncology, Xiangya HospitalCentral South UniversityHunanChina
| | - Changhao Huang
- Department of Oncology, Xiangya HospitalCentral South UniversityHunanChina
| | - Shangwei Zhong
- The Cancer Research Institute, Hengyang Medical SchoolUniversity of South ChinaHengyangChina
| |
Collapse
|
5
|
Castillo-Castrejon M, Sankofi BM, Murguia SJ, Udeme AA, Cen HH, Xia YH, Thomas NS, Berry WL, Jones KL, Richard VR, Zahedi RP, Borchers CH, Johnson JD, Wellberg EA. FGF1 supports glycolytic metabolism through the estrogen receptor in endocrine-resistant and obesity-associated breast cancer. Breast Cancer Res 2023; 25:99. [PMID: 37608351 PMCID: PMC10463730 DOI: 10.1186/s13058-023-01699-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/16/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Obesity increases breast cancer risk and breast cancer-specific mortality, particularly for people with estrogen receptor (ER)-positive tumors. Body mass index (BMI) is used to define obesity, but it may not be the best predictor of breast cancer risk or prognosis on an individual level. Adult weight gain is an independent indicator of breast cancer risk. Our previous work described a murine model of obesity, ER-positive breast cancer, and weight gain and identified fibroblast growth factor receptor (FGFR) as a potential driver of tumor progression. During adipose tissue expansion, the FGF1 ligand is produced by hypertrophic adipocytes as a stimulus to stromal preadipocytes that proliferate and differentiate to provide additional lipid storage capacity. In breast adipose tissue, FGF1 production may stimulate cancer cell proliferation and tumor progression. METHODS We explored the effects of FGF1 on ER-positive endocrine-sensitive and resistant breast cancer and compared that to the effects of the canonical ER ligand, estradiol. We used untargeted proteomics, specific immunoblot assays, gene expression profiling, and functional metabolic assessments of breast cancer cells. The results were validated in tumors from obese mice and breast cancer datasets from women with obesity. RESULTS FGF1 stimulated ER phosphorylation independently of estradiol in cells that grow in obese female mice after estrogen deprivation treatment. Phospho- and total proteomic, genomic, and functional analyses of endocrine-sensitive and resistant breast cancer cells show that FGF1 promoted a cellular phenotype characterized by glycolytic metabolism. In endocrine-sensitive but not endocrine-resistant breast cancer cells, mitochondrial metabolism was also regulated by FGF1. Comparison of gene expression profiles indicated that tumors from women with obesity shared hallmarks with endocrine-resistant breast cancer cells. CONCLUSIONS Collectively, our data suggest that one mechanism by which obesity and weight gain promote breast cancer progression is through estrogen-independent ER activation and cancer cell metabolic reprogramming, partly driven by FGF/FGFR. The first-line treatment for many patients with ER-positive breast cancer is inhibition of estrogen synthesis using aromatase inhibitors. In women with obesity who are experiencing weight gain, locally produced FGF1 may activate ER to promote cancer cell metabolic reprogramming and tumor progression independently of estrogen.
Collapse
Affiliation(s)
- Marisol Castillo-Castrejon
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA
| | - Barbara Mensah Sankofi
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA
| | - Stevi Johnson Murguia
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA
| | - Abasi-Ama Udeme
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA
| | - Hoaning Howard Cen
- Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Yi Han Xia
- Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Nisha S Thomas
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA
| | - William L Berry
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA
| | - Kenneth L Jones
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA
| | - Vincent R Richard
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital and McGill University, Montreal, QC, Canada
| | - Rene P Zahedi
- Manitoba Centre for Proteomics and Systems Biology, Winnipeg, MB, R3E 3P4, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB, R3E 3P4, Canada
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada
- CancerCare Manitoba Research Institute, Winnipeg, MB, R3E 0V9, Canada
| | - Christoph H Borchers
- Manitoba Centre for Proteomics and Systems Biology, Winnipeg, MB, R3E 3P4, Canada
- Gerald Bronfman Department of Oncology, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, H3T 1E2, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Montreal, QC, H3A 2B4, Canada
| | - James D Johnson
- Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Elizabeth A Wellberg
- Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
6
|
Nguyen HL, Geukens T, Maetens M, Aparicio S, Bassez A, Borg A, Brock J, Broeks A, Caldas C, Cardoso F, De Schepper M, Delorenzi M, Drukker CA, Glas AM, Green AR, Isnaldi E, Eyfjörð J, Khout H, Knappskog S, Krishnamurthy S, Lakhani SR, Langerod A, Martens JWM, McCart Reed AE, Murphy L, Naulaerts S, Nik-Zainal S, Nevelsteen I, Neven P, Piccart M, Poncet C, Punie K, Purdie C, Rakha EA, Richardson A, Rutgers E, Vincent-Salomon A, Simpson PT, Schmidt MK, Sotiriou C, Span PN, Tan KTB, Thompson A, Tommasi S, Van Baelen K, Van de Vijver M, Van Laere S, Van't Veer L, Viale G, Viari A, Vos H, Witteveen AT, Wildiers H, Floris G, Garg AD, Smeets A, Lambrechts D, Biganzoli E, Richard F, Desmedt C. Obesity-associated changes in molecular biology of primary breast cancer. Nat Commun 2023; 14:4418. [PMID: 37479706 PMCID: PMC10361985 DOI: 10.1038/s41467-023-39996-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 07/06/2023] [Indexed: 07/23/2023] Open
Abstract
Obesity is associated with an increased risk of developing breast cancer (BC) and worse prognosis in BC patients, yet its impact on BC biology remains understudied in humans. This study investigates how the biology of untreated primary BC differs according to patients' body mass index (BMI) using data from >2,000 patients. We identify several genomic alterations that are differentially prevalent in overweight or obese patients compared to lean patients. We report evidence supporting an ageing accelerating effect of obesity at the genetic level. We show that BMI-associated differences in bulk transcriptomic profile are subtle, while single cell profiling allows detection of more pronounced changes in different cell compartments. These analyses further reveal an elevated and unresolved inflammation of the BC tumor microenvironment associated with obesity, with distinct characteristics contingent on the estrogen receptor status. Collectively, our analyses imply that obesity is associated with an inflammaging-like phenotype. We conclude that patient adiposity may play a significant role in the heterogeneity of BC and should be considered for BC treatment tailoring.
Collapse
Affiliation(s)
- Ha-Linh Nguyen
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Tatjana Geukens
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Marion Maetens
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Samuel Aparicio
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Ayse Bassez
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Ake Borg
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden
- Lund University Cancer Center Lund, Lund, Sweden
- CREATE Health Strategic Centre for Translational Cancer Research, Lund University, Lund, Sweden
- Department of Clinical Sciences, SCIBLU Genomics, Lund University, Lund, Sweden
| | - Jane Brock
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Annegien Broeks
- Departments of Core Facility, Molecular Pathology and Biobanking, Antoni van Leeuwenhoek, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Fatima Cardoso
- Breast Unit, Champalimaud Clinical Center/Champalimaud Foundation, Lisbon, Portugal
| | - Maxim De Schepper
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Mauro Delorenzi
- Department of Oncology, University of Lausanne, Epalinges, Switzerland
- SIB Swiss Institute of Bioinformatics, Bioinformatics Core Facility, Lausanne, Switzerland
| | - Caroline A Drukker
- Department of Surgical Oncology, Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | | | - Andrew R Green
- Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Edoardo Isnaldi
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jórunn Eyfjörð
- BioMedical Center, School of Health Sciences, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Hazem Khout
- Department of Breast Surgery, Glenfield Hospital, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Stian Knappskog
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Savitri Krishnamurthy
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sunil R Lakhani
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
- Pathology Queensland, The Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Anita Langerod
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Ullernchausseen, Oslo, Norway
| | - John W M Martens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Amy E McCart Reed
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Leigh Murphy
- University of Manitoba and Cancer Care Manitoba Research Institute, Winnipeg, MB, Canada
| | - Stefan Naulaerts
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Serena Nik-Zainal
- Department of Medical Genetics, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- MRC Cancer Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Ines Nevelsteen
- Department of Surgical Oncology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Patrick Neven
- Department of Gynecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Martine Piccart
- Institut Jules Bordet and Université Libre de Bruxelles, Brussels, Belgium
| | - Coralie Poncet
- European Organisation for Research and Treatment of Cancer (EORTC) Headquarters, Brussels, Belgium
| | - Kevin Punie
- Department of General Medical Oncology and Multidisciplinary Breast Unit, Leuven Cancer Institute and University Hospitals Leuven, Leuven, Belgium
| | - Colin Purdie
- Department of Pathology, University of Dundee, NHS Tayside, Dundee, UK
| | - Emad A Rakha
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Histopathology, Nottingham University Hospital NHS Trust, City Hospital Campus, Nottingham, UK
| | | | - Emiel Rutgers
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Anne Vincent-Salomon
- Diagnostic and Theranostic Medicine Division, Institut Curie, PSL Research University, Paris, France
| | - Peter T Simpson
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Marjanka K Schmidt
- Division of Molecular Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Christos Sotiriou
- Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium
| | - Paul N Span
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Kiat Tee Benita Tan
- Department of General Surgery, Sengkang General Hospital, Singapore, Singapore
- Department of Breast Surgery, Singapore General Hospital, Singapore, Singapore
- Department of Breast Surgery, National Cancer Centre, Singapore, Singapore
| | - Alastair Thompson
- Department of Surgery, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Stefania Tommasi
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumouri "Giovanni Paolo II", Bari, Italy
| | - Karen Van Baelen
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Marc Van de Vijver
- Department of Pathology, Amsterdam University Medical Centers, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Steven Van Laere
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Laura Van't Veer
- Department of Laboratory Medicine, UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Giuseppe Viale
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Alain Viari
- Synergie Lyon Cancer, Plateforme de Bio-informatique 'Gilles Thomas', Lyon, France
| | - Hanne Vos
- Department of Surgical Oncology, University Hospitals Leuven, Leuven, Belgium
| | | | - Hans Wildiers
- Department of General Medical Oncology and Multidisciplinary Breast Unit, Leuven Cancer Institute and University Hospitals Leuven, Leuven, Belgium
| | - Giuseppe Floris
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ann Smeets
- Department of Surgical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Elia Biganzoli
- Unit of Medical Statistics, Biometry and Epidemiology, Department of Biomedical and Clinical Sciences (DIBIC) "L. Sacco" & DSRC, LITA Vialba campus, Università degli Studi di Milano, Milan, Italy
| | - François Richard
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Christine Desmedt
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
7
|
Rosario SR, Dong B, Zhang Y, Hsiao HH, Isenhart E, Wang J, Siegel EM, Monjazeb AM, Owen DH, Dey P, Tabung FK, Spakowicz DJ, Murphy WJ, Edge S, Yendamuri S, Ibrahimi S, Kolesar JM, McDonald PH, Vadehra D, Churchman M, Liu S, Kalinski P, Mukherjee S. Metabolic Dysregulation Explains the Diverse Impacts of Obesity in Males and Females with Gastrointestinal Cancers. Int J Mol Sci 2023; 24:10847. [PMID: 37446025 PMCID: PMC10342094 DOI: 10.3390/ijms241310847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/09/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
The prevalence of obesity, defined as the body mass index (BMI) ≥ 30 kg/m2, has reached epidemic levels. Obesity is associated with an increased risk of various cancers, including gastrointestinal ones. Recent evidence has suggested that obesity disproportionately impacts males and females with cancer, resulting in varied transcriptional and metabolic dysregulation. This study aimed to elucidate the differences in the metabolic milieu of adenocarcinomas of the gastrointestinal (GI) tract both related and unrelated to sex in obesity. To demonstrate these obesity and sex-related effects, we utilized three primary data sources: serum metabolomics from obese and non-obese patients assessed via the Biocrates MxP Quant 500 mass spectrometry-based kit, the ORIEN tumor RNA-sequencing data for all adenocarcinoma cases to assess the impacts of obesity, and publicly available TCGA transcriptional analysis to assess GI cancers and sex-related differences in GI cancers specifically. We applied and integrated our unique transcriptional metabolic pipeline in combination with our metabolomics data to reveal how obesity and sex can dictate differential metabolism in patients. Differentially expressed genes (DEG) analysis of ORIEN obese adenocarcinoma as compared to normal-weight adenocarcinoma patients resulted in large-scale transcriptional reprogramming (4029 DEGs, adj. p < 0.05 and |logFC| > 0.58). Gene Set Enrichment and metabolic pipeline analysis showed genes enriched for pathways relating to immunity (inflammation, and CD40 signaling, among others) and metabolism. Specifically, we found alterations to steroid metabolism and tryptophan/kynurenine metabolism in obese patients, both of which are highly associated with disease severity and immune cell dysfunction. These findings were further confirmed using the TCGA colorectal adenocarcinoma (CRC) and esophageal adenocarcinoma (ESCA) data, which showed similar patterns of increased tryptophan catabolism for kynurenine production in obese patients. These patients further showed disparate alterations between males and females when comparing obese to non-obese patient populations. Alterations to immune and metabolic pathways were validated in six patients (two obese and four normal weight) via CD8+/CD4+ peripheral blood mononuclear cell RNA-sequencing and paired serum metabolomics, which showed differential kynurenine and lipid metabolism, which corresponded with altered T-cell transcriptome in obese populations. Overall, obesity is associated with differential transcriptional and metabolic programs in various disease sites. Further, these alterations, such as kynurenine and tryptophan metabolism, which impact both metabolism and immune phenotype, vary with sex and obesity together. This study warrants further in-depth investigation into obesity and sex-related alterations in cancers that may better define biomarkers of response to immunotherapy.
Collapse
Affiliation(s)
- Spencer R. Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (S.R.R.); (Y.Z.); (H.-H.H.); (E.I.); (J.W.); (S.L.)
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Bowen Dong
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (B.D.); (P.D.); (P.K.)
| | - Yali Zhang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (S.R.R.); (Y.Z.); (H.-H.H.); (E.I.); (J.W.); (S.L.)
| | - Hua-Hsin Hsiao
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (S.R.R.); (Y.Z.); (H.-H.H.); (E.I.); (J.W.); (S.L.)
| | - Emily Isenhart
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (S.R.R.); (Y.Z.); (H.-H.H.); (E.I.); (J.W.); (S.L.)
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (S.R.R.); (Y.Z.); (H.-H.H.); (E.I.); (J.W.); (S.L.)
| | - Erin M. Siegel
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Arta M. Monjazeb
- Department of Radiation Oncology, University of California Davis, Sacramento, CA 95616, USA;
| | - Dwight H. Owen
- Department of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (D.H.O.); (D.J.S.)
| | - Prasenjit Dey
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (B.D.); (P.D.); (P.K.)
| | - Fred K. Tabung
- Department of Epidemiology, Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA;
| | - Daniel J. Spakowicz
- Department of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (D.H.O.); (D.J.S.)
| | - William J. Murphy
- Department of Immunology, University of California Davis, Sacramento, CA 95616, USA;
| | - Stephen Edge
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA;
| | - Sai Yendamuri
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA;
| | - Sami Ibrahimi
- Department of Medicine, Oklahoma University Health Stephenson Cancer Center, Oklahoma City, OK 73104, USA;
| | - Jill M. Kolesar
- Department of Pharmacy, University of Kentucky College of Pharmacy, Lexington, KY 40506, USA;
| | - Patsy H. McDonald
- Department of Cancer Biology, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Deepak Vadehra
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA;
| | - Michelle Churchman
- Precision Therapy and Diagnostics, Aster Insights, Hudson, FL 34667, USA;
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (S.R.R.); (Y.Z.); (H.-H.H.); (E.I.); (J.W.); (S.L.)
| | - Pawel Kalinski
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (B.D.); (P.D.); (P.K.)
| | - Sarbajit Mukherjee
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (B.D.); (P.D.); (P.K.)
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA;
| |
Collapse
|
8
|
Zhu J, Min N, Gong W, Chen Y, Li X. Identification of Hub Genes and Biological Mechanisms Associated with Non-Alcoholic Fatty Liver Disease and Triple-Negative Breast Cancer. Life (Basel) 2023; 13:life13040998. [PMID: 37109526 PMCID: PMC10146727 DOI: 10.3390/life13040998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/20/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
The relationship between non-alcoholic fatty liver disease (NAFLD) and triple-negative breast cancer (TNBC) has been widely recognized, but the underlying mechanisms are still unknown. The objective of this study was to identify the hub genes associated with NAFLD and TNBC, and to explore the potential co-pathogenesis and prognostic linkage of these two diseases. We used GEO, TCGA, STRING, ssGSEA, and Rstudio to investigate the common differentially expressed genes (DEGs), conduct functional and signaling pathway enrichment analyses, and determine prognostic value between TNBC and NAFLD. GO and KEGG enrichment analyses of the common DEGs showed that they were enriched in leukocyte aggregation, migration and adhesion, apoptosis regulation, and the PPAR signaling pathway. Fourteen candidate hub genes most likely to mediate NAFLD and TNBC occurrence were identified and validation results in a new cohort showed that ITGB2, RAC2, ITGAM, and CYBA were upregulated in both diseases. A univariate Cox analysis suggested that high expression levels of ITGB2, RAC2, ITGAM, and CXCL10 were associated with a good prognosis in TNBC. Immune infiltration analysis of TNBC samples showed that NCF2, ICAM1, and CXCL10 were significantly associated with activated CD8 T cells and activated CD4 T cells. NCF2, CXCL10, and CYBB were correlated with regulatory T cells and myeloid-derived suppressor cells. This study demonstrated that the redox reactions regulated by the NADPH oxidase (NOX) subunit genes and the transport and activation of immune cells regulated by integrins may play a central role in the co-occurrence trend of NAFLD and TNBC. Additionally, ITGB2, RAC2, and ITGAM were upregulated in both diseases and were prognostic protective factors of TNBC; they may be potential therapeutic targets for treatment of TNBC patients with NAFLD, but further experimental studies are still needed.
Collapse
Affiliation(s)
- Jingjin Zhu
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Ningning Min
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Wenye Gong
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
- Medical School of Chinese PLA, Beijing 100853, China
| | - Yizhu Chen
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
- Medical School of Chinese PLA, Beijing 100853, China
| | - Xiru Li
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
9
|
Min J, Yeon S, Ryu J, Kim JY, Yang EJ, Kim SI, Park S, Jeon JY. Shoulder function and health outcomes in newly diagnosed breast cancer patients receiving surgery: a prospective study. Clin Breast Cancer 2023; 23:e247-e258. [PMID: 36990840 DOI: 10.1016/j.clbc.2023.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND Breast cancer surgery is associated with upper-body morbidities that may last several years postsurgery. Research has not determined if the type of surgery leads to differential effects on shoulder function, activity levels, and QoL during the early rehabilitation period. The main objective of this study is to examine changes in shoulder function, health, and fitness outcomes from the day before to surgery to 6 months postsurgery. PATIENTS AND METHODS We recruited breast cancer patients (N = 70) scheduled to receive breast cancer surgery at Severance Hospital in Seoul to participate in this prospective study. Shoulder range of motion (ROM) and upper body strength, the disabilities of Arm, Shoulder, and Hand (quick-DASH), body composition, physical activity levels, and QoL were measured at baseline (presurgery) and then weekly for 4 weeks, and at 3 months and 6 months postsurgery. RESULTS During 6 months after surgery, shoulder ROM was reduced only affected arm while shoulder strength was significantly declined in both affected and unaffected arms. Within 4 weeks postsurgery, patients who underwent total mastectomy were significantly less recovered than patients with partial mastectomy in ROM of flexion (P < .05) and abduction (P < .05). However, shoulder strength of both arms observed no interaction between surgical type and time. We observed significant changes in body composition, quick-DASH score, physical activity levels, and QoL from presurgery to 6-months postsurgery. CONCLUSIONS Shoulder function, activity levels, and QoL improved significantly from surgery to 6-months postsurgery. Surgery type influenced changes in shoulder ROM.
Collapse
|
10
|
Molania R, Foroutan M, Gagnon-Bartsch JA, Gandolfo LC, Jain A, Sinha A, Olshansky G, Dobrovic A, Papenfuss AT, Speed TP. Removing unwanted variation from large-scale RNA sequencing data with PRPS. Nat Biotechnol 2023; 41:82-95. [PMID: 36109686 PMCID: PMC9849124 DOI: 10.1038/s41587-022-01440-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 06/30/2022] [Indexed: 01/22/2023]
Abstract
Accurate identification and effective removal of unwanted variation is essential to derive meaningful biological results from RNA sequencing (RNA-seq) data, especially when the data come from large and complex studies. Using RNA-seq data from The Cancer Genome Atlas (TCGA), we examined several sources of unwanted variation and demonstrate here how these can significantly compromise various downstream analyses, including cancer subtype identification, association between gene expression and survival outcomes and gene co-expression analysis. We propose a strategy, called pseudo-replicates of pseudo-samples (PRPS), for deploying our recently developed normalization method, called removing unwanted variation III (RUV-III), to remove the variation caused by library size, tumor purity and batch effects in TCGA RNA-seq data. We illustrate the value of our approach by comparing it to the standard TCGA normalizations on several TCGA RNA-seq datasets. RUV-III with PRPS can be used to integrate and normalize other large transcriptomic datasets coming from multiple laboratories or platforms.
Collapse
Affiliation(s)
- Ramyar Molania
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Momeneh Foroutan
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | | | - Luke C Gandolfo
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Aryan Jain
- Department of Economics and Statistics, Monash University, Melbourne, Victoria, Australia
| | - Abhishek Sinha
- Department of Economics and Statistics, Monash University, Melbourne, Victoria, Australia
| | - Gavriel Olshansky
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Alexander Dobrovic
- Department of Surgery, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia
| | - Anthony T Papenfuss
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia.
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Terence P Speed
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
11
|
Ilozumba MN, Yao S, Llanos AAM, Omilian AR, Zhang W, Datta S, Hong CC, Davis W, Khoury T, Bandera EV, Higgins M, Ambrosone CB, Cheng TYD. mTOR pathway gene expression in association with race and clinicopathological characteristics in Black and White breast cancer patients. Discov Oncol 2022; 13:34. [PMID: 35608730 PMCID: PMC9130392 DOI: 10.1007/s12672-022-00497-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/13/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Aberrant activation of the mammalian Target of Rapamycin (mTOR) pathway has been linked to obesity and endocrine therapy resistance, factors that may contribute to Black-White disparities in breast cancer outcomes. We evaluated associations of race and clinicopathological characteristics with mRNA expression of key mTOR pathway genes in breast tumors. METHODS Surgical tumor tissue blocks were collected from 367 newly diagnosed breast cancer patients (190 Black and 177 White). Gene expression of AKT1, EIF4EBP1, MTOR, RPS6KB2, and TSC1 were quantified by NanoString nCounter. Differential gene expression was assessed using linear regression on log2-transformed values. Gene expression and DNA methylation data from TCGA were used for validation and investigation of race-related differences. RESULTS Compared to White women, Black women had relative under-expression of AKT1 (log2 fold-change = - 0.31, 95% CI - 0.44, - 0.18) and RPS6KB2 (log2 fold-change = - 0.11, 95% CI - 0.19, - 0.03). Higher vs. lower tumor grade was associated with relative over-expression of EIF4EBP1 and RPS6KB2, but with lower expression of TSC1. Compared to luminal tumors, triple-negative tumors had relative under-expression of TSC1 (log2 fold-change = - 0.42, 95% CI - 0.22, - 0.01). The results were similar in the TCGA breast cancer dataset. Post-hoc analyses identified differential CpG methylation within the AKT1 and RPS6KB2 locus between Black and White women. CONCLUSIONS Over-expression of RPS6KB2 and EIF4EBP1 and under-expression of TSC1 might be indicators of more aggressive breast cancer phenotypes. Differential expression of AKT1 and RPS6KB2 by race warrants further investigation to elucidate their roles in racial disparities of treatment resistance and outcomes between Black and White women with breast cancer.
Collapse
Affiliation(s)
| | - Song Yao
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Adana A M Llanos
- Department of Epidemiology, Mailman School of Public Health and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, NY, New York, United States
| | - Angela R Omilian
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Susmita Datta
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Chi-Chen Hong
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Warren Davis
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Thaer Khoury
- Department of Pathology & Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Elisa V Bandera
- Cancer Epidemiology and Health Outcomes, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Michael Higgins
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Christine B Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Ting-Yuan David Cheng
- Department of Epidemiology, University of Florida, Gainesville, FL, USA.
- Division of Cancer Prevention and Control, Department of Internal Medicine, The Ohio State University, Suite 525, 1590 North High Street, Columbus, OH, 43201, USA.
| |
Collapse
|
12
|
Hong C, Yan Y, Su L, Chen D, Zhang C. Development of a risk-stratification scoring system for predicting risk of breast cancer based on non-alcoholic fatty liver disease, non-alcoholic fatty pancreas disease, and uric acid. Open Med (Wars) 2022; 17:619-625. [PMID: 35434374 PMCID: PMC8974397 DOI: 10.1515/med-2022-0462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 02/15/2022] [Accepted: 02/22/2022] [Indexed: 12/24/2022] Open
Abstract
Many breast cancer patients have both non-alcoholic fatty liver disease (NAFLD) and non-alcoholic fatty pancreas disease (NAFPD). Consequently, we hypothesized that NAFPD and NAFLD were associated with breast cancer, and aimed to build a novel risk-stratification scoring system based on it. In this study, a total of 961 patients with breast cancer and 1,006 non-cancer patients were recruited. The clinical characteristics were collected and analyzed using logistic analysis. Risk factors were assessed by a risk rating system. Univariate analysis showed that body mass index, triglyceride, total cholesterol, NAFLD, NAFPD, low-density lipoprotein, and uric acid (UA) were significantly related to breast cancer. Among them, NAFLD, NAFPD, and UA were independent risk factors related to breast cancer identified by multivariate analysis. The risk assessment model was established based on these factors and demonstrated that the odds ratio sharply increased with the rising scores. Compared with the low-risk group, the odds ratio in the intermediate- and high-risk groups were 1.662 (1.380–2.001) and 3.185 (2.145–4.728), respectively. In conclusion, the risk-stratification scoring system combining NAFLD, NAFPD, and UA can accurately predict the occurrence of breast cancer.
Collapse
Affiliation(s)
- Chuntian Hong
- Department of Ultrasound, Quanzhou First Hospital Affiliated to Fujian Medical University , Quanzhou 362000 , China
| | - Yonghao Yan
- Department of Ultrasound, Quanzhou First Hospital Affiliated to Fujian Medical University , Quanzhou 362000 , China
| | - Liyang Su
- Department of Ultrasound, Quanzhou First Hospital Affiliated to Fujian Medical University , Quanzhou 362000 , China
| | - Debo Chen
- Department of Breast Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University , Quanzhou 362000 , China
| | - Changqing Zhang
- Department of Gastroenterology, Quanzhou First Hospital Affiliated to Fujian Medical University , Quanzhou 362000 , China
| |
Collapse
|
13
|
Argyrakopoulou G, Dalamaga M, Spyrou N, Kokkinos A. Gender Differences in Obesity-Related Cancers. Curr Obes Rep 2021; 10:100-115. [PMID: 33523397 DOI: 10.1007/s13679-021-00426-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/11/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW In this review, we summarize the role of obesity in carcinogenesis, providing details on specific cancer sites. Special emphasis is given to gender differences in obesity-related cancers and on the effect of bariatric surgery on cancer risk. RECENT FINDINGS Accumulating evidence has highlighted the detrimental role of overweight/obesity in cancer, with almost 55% of cancers diagnosed in women and 24% diagnosed in men considered overweight- and obesity-related cancers. Sufficient data have shown that higher BMI is associated with risk of gynecologic malignancies (mainly breast and endometrial cancers) as well as cancers in sites such as the esophagus (adenocarcinoma), gastric cardia, colon, rectum, liver, gallbladder, pancreas, kidney, thyroid gland, and multiple myeloma. The main mechanisms underlying this relationship include the insulin/IGF1 system, the effect of sex hormones, and adipocytokines. Marked differences may be seen in specific cancer sites when comparing men to women. There is a higher overall incidence of obesity-related cancers among females (endometrial, ovarian, and postmenopausal female breast cancers), whereas cancers that concern both sexes show a higher incidence in males, particularly esophageal adenocarcinoma (male to female ratio: 9: 1 in the USA). Additionally, bariatric surgery has provided evidence of lowering overall cancer risk in patients with morbid obesity. Interestingly, bariatric surgery may lower overall cancer risk in women within the first 5 years after surgery due to the reduced risk of breast and endometrial cancer, and non-Hodgkin lymphoma. Obesity constitutes the base for marked metabolic, hormonal, and inflammatory alterations, including increased cancer risk in both men and women. Implementation of early obesity prevention strategies could ameliorate the continuously increasing incidence of cancer attributed to obesity.
Collapse
Affiliation(s)
- Georgia Argyrakopoulou
- Diabetes and Obesity Unit, Athens Medical Center, Distomou 5-7, Amaroussio, 15127, Athens, Greece.
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece
| | - Nikolaos Spyrou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece
| | - Alexander Kokkinos
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko Hospital, 115 27, Athens, Greece
| |
Collapse
|
14
|
Cholesterol-Induced Metabolic Reprogramming in Breast Cancer Cells Is Mediated via the ERRα Pathway. Cancers (Basel) 2021; 13:cancers13112605. [PMID: 34073320 PMCID: PMC8198778 DOI: 10.3390/cancers13112605] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/08/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary There is increasing evidence that obesity and high circulating cholesterol levels are associated with an increased risk of recurrence and a higher mortality rate in breast cancer patients via altering the metabolic programming in breast cancer cells. However, the underlying molecular mechanism by which high cholesterol levels reprogram the metabolic pathways in breast cancer cells is not well-understood. We have previously demonstrated that cholesterol acts as an endogenous agonist of estrogen-related receptor α (ERRα), a strong regulator of cellular metabolism. The aim of the current study is to demonstrate whether cholesterol/obesity mediates its pathogenic effect in breast cancer cells via altering metabolic pathways in an ERRα-dependent manner. The findings of this study provide mechanistic insights into the link between cholesterol/obesity and metabolic reprogramming in breast cancer patients and reveal the metabolic vulnerabilities in such breast cancer patients that could be therapeutically targeted. Abstract The molecular mechanism underlying the metabolic reprogramming associated with obesity and high blood cholesterol levels is poorly understood. We previously reported that cholesterol is an endogenous ligand of the estrogen-related receptor alpha (ERRα). Using functional assays, metabolomics, and genomics, here we show that exogenous cholesterol alters the metabolic pathways in estrogen receptor-positive (ER+) and triple-negative breast cancer (TNBC) cells, and that this involves increased oxidative phosphorylation (OXPHOS) and TCA cycle intermediate levels. In addition, cholesterol augments aerobic glycolysis in TNBC cells although it remains unaltered in ER+ cells. Interestingly, cholesterol does not alter the metabolite levels of glutaminolysis, one-carbon metabolism, or the pentose phosphate pathway, but increases the NADPH levels and cellular proliferation, in both cell types. Importantly, we show that the above cholesterol-induced modulations of the metabolic pathways in breast cancer cells are mediated via ERRα. Furthermore, analysis of the ERRα metabolic gene signature of basal-like breast tumours of overweight/obese versus lean patients, using the GEO database, shows that obesity may modulate ERRα gene signature in a manner consistent with our in vitro findings with exogenous cholesterol. Given the close link between high cholesterol levels and obesity, our findings provide a mechanistic explanation for the association between cholesterol/obesity and metabolic reprogramming in breast cancer patients.
Collapse
|
15
|
Maguire OA, Ackerman SE, Szwed SK, Maganti AV, Marchildon F, Huang X, Kramer DJ, Rosas-Villegas A, Gelfer RG, Turner LE, Ceballos V, Hejazi A, Samborska B, Rahbani JF, Dykstra CB, Annis MG, Luo JD, Carroll TS, Jiang CS, Dannenberg AJ, Siegel PM, Tersey SA, Mirmira RG, Kazak L, Cohen P. Creatine-mediated crosstalk between adipocytes and cancer cells regulates obesity-driven breast cancer. Cell Metab 2021; 33:499-512.e6. [PMID: 33596409 PMCID: PMC7954401 DOI: 10.1016/j.cmet.2021.01.018] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 11/24/2020] [Accepted: 01/27/2021] [Indexed: 01/08/2023]
Abstract
Obesity is a major risk factor for adverse outcomes in breast cancer; however, the underlying molecular mechanisms have not been elucidated. To investigate the role of crosstalk between mammary adipocytes and neoplastic cells in the tumor microenvironment (TME), we performed transcriptomic analysis of cancer cells and adjacent adipose tissue in a murine model of obesity-accelerated breast cancer and identified glycine amidinotransferase (Gatm) in adipocytes and Acsbg1 in cancer cells as required for obesity-driven tumor progression. Gatm is the rate-limiting enzyme in creatine biosynthesis, and deletion in adipocytes attenuated obesity-driven tumor growth. Similarly, genetic inhibition of creatine import into cancer cells reduced tumor growth in obesity. In parallel, breast cancer cells in obese animals upregulated the fatty acyl-CoA synthetase Acsbg1 to promote creatine-dependent tumor progression. These findings reveal key nodes in the crosstalk between adipocytes and cancer cells in the TME necessary for obesity-driven breast cancer progression.
Collapse
Affiliation(s)
- Olivia A Maguire
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY 10065, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-institutional MD-PhD Program, New York, NY 10065, USA
| | - Sarah E Ackerman
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY 10065, USA; AAAS Science and Technology Policy Fellow in the Office of Global Health, Health Workforce Branch, U.S. Agency for International Development, Washington, D.C. 20547, USA
| | - Sarah K Szwed
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY 10065, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-institutional MD-PhD Program, New York, NY 10065, USA
| | - Aarthi V Maganti
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY 10065, USA
| | - François Marchildon
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY 10065, USA
| | - Xiaojing Huang
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY 10065, USA; Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Daniel J Kramer
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY 10065, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-institutional MD-PhD Program, New York, NY 10065, USA
| | | | - Rebecca G Gelfer
- Weill Cornell/Rockefeller/Sloan Kettering Tri-institutional MD-PhD Program, New York, NY 10065, USA
| | - Lauren E Turner
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY 10065, USA
| | - Victor Ceballos
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY 10065, USA
| | - Asal Hejazi
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY 10065, USA
| | - Bozena Samborska
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G1Y6, Canada
| | - Janane F Rahbani
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G1Y6, Canada
| | - Christien B Dykstra
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G1Y6, Canada
| | - Matthew G Annis
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Ji-Dung Luo
- Bioinformatics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Thomas S Carroll
- Bioinformatics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Caroline S Jiang
- Rockefeller University Hospital, The Rockefeller University, New York, NY 10065, USA
| | - Andrew J Dannenberg
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Peter M Siegel
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G1Y6, Canada
| | - Sarah A Tersey
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | | | - Lawrence Kazak
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G1Y6, Canada
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
16
|
Jiang J, Guo Z, Xu J, Sun T, Zheng X. Identification of Aurora Kinase A as a Biomarker for Prognosis in Obesity Patients with Early Breast Cancer. Onco Targets Ther 2020; 13:4971-4985. [PMID: 32581556 PMCID: PMC7276210 DOI: 10.2147/ott.s250619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/25/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Obesity is associated both with a higher risk of developing breast cancer, particularly in postmenopausal women, and with worse disease outcome for women of all ages. Previous investigation suggested Aurora A kinase was able to partially restore the functionalities of obese adipose-derived mesenchymal stem cells by stabilizing their primary cilia and reestablishing a balance of multiple stemness-associated genes. The association between Aurora A and obesity breast cancer is still unclear. We hypothesized that overexpression of Aurora A was associated with poor survival in obesity breast cancer and the related axis mechanism was involved. METHODS A total of 517 primary breast cancer specimens were collected from the First Affiliated Hospital of China Medical University between January 2011 and November 2016. Our independent variable was BMI at baseline, categorized as overweight (BMI ≥25 kg/m2, as obesity cohort), and normal (18.5 ≤ BMI <25 kg/m2, as non-obesity cohort). The immunohistochemical (IHC) staining was performed with Aurora A, Survivin, MMP11, Cyclin B1, and Cathepsin L. Kaplan-Meier curve was used to analyze overall survival in our cohorts and TCGA-BRCA data (GSE3494). Log rank test was used to calculate P values. Protein-protein interaction (PPI) network analysis and MCODE model were used to analyze the Aurora-altered signal pathway from GSE78958. RESULTS Among 517 breast patients, Aurora A-positive (staining scores ≥4) was significantly higher in obesity breast carcinoma compared with non-obesity cancer carcinoma (χ 2=9.79, P=0.002), with more frequency in hormone receptor-negative (68.4% vs 77.9%, P=0.015) and HER2-positive patients (28.7% vs 17.9%, P=0.003). High Aurora A expression was remarkably and significantly associated with overall survival (OS) (8-year OS ratio: 69.5% vs 81.1%, OR=1.76, 95% CI: 1.03~3.02, P=0.041) in obesity cohort. Interestingly, higher expression of Aurora A was not associated with a shorter overall survival time among the non-obesity breast cancer (8-year OS ratio: 81.4% vs 85.8%, OR=1.40, 95% CI: 0.79~2.45, P=0.229). As for RFS, the expression levels of Aurora A expression genes have no significance with RFS statistically in non-obesity and obesity patients. Aurora A and lymph node metastases were significantly poor prognostic factors for OS, and borderline significance was noted for high BMI. Kaplan-Meier survival analysis from TCGA database confirmed that the high Aurora A expression group had worse prognosis (HR=1.47, 95% CI: 1.14-1.90, P=0.003). The KEGG pathway enrichment results were consistent with GO biological process term analysis, in which CCNB1 was enriched for upregulated Aurora A. In our samples, Aurora A level on tumor cytoplasm had broad connections with Cyclin B1 by IHC correlation analysis (correlation coefficient = 0.227, P=0.001). CONCLUSION Our finding demonstrates here for the first time that high expression of Aurora A was notably correlated with early recurrence and poor overall survival in obesity patients with early breast cancer. The Aurora A-Cyclin B1 axis could be a potential promising therapeutic target for cancer intervention and therapy.
Collapse
Affiliation(s)
- Junhan Jiang
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Zihe Guo
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Junnan Xu
- Department of Breast Medical, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, People’s Republic of China
| | - Tao Sun
- Department of Breast Medical, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, People’s Republic of China
| | - Xinyu Zheng
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
- Laboratory 1, Cancer Institute, The First Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
17
|
Hillers-Ziemer LE, Arendt LM. Weighing the Risk: effects of Obesity on the Mammary Gland and Breast Cancer Risk. J Mammary Gland Biol Neoplasia 2020; 25:115-131. [PMID: 32519090 PMCID: PMC7933979 DOI: 10.1007/s10911-020-09452-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022] Open
Abstract
Obesity is a preventable risk factor for breast cancer following menopause. Regardless of menopausal status, obese women who develop breast cancer have a worsened prognosis. Breast tissue is comprised of mammary epithelial cells organized into ducts and lobules and surrounded by adipose-rich connective tissue. Studies utilizing multiple in vivo models of obesity as well as human breast tissue have contributed to our understanding of how obesity alters mammary tissue. Localized changes in mammary epithelial cell populations, elevated secretion of adipokines and angiogenic mediators, inflammation within mammary adipose tissue, and remodeling of the extracellular matrix may result in an environment conducive to breast cancer growth. Despite these significant alterations caused by obesity within breast tissue, studies have suggested that some, but not all, obesity-induced changes may be mitigated with weight loss. Here, we review our current understanding regarding the impact of obesity on the breast microenvironment, how obesity-induced changes may contribute to breast tumor progression, and the impact of weight loss on the breast microenvironment.
Collapse
Affiliation(s)
- Lauren E Hillers-Ziemer
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI, 53706, USA
| | - Lisa M Arendt
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI, 53706, USA.
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA.
| |
Collapse
|
18
|
Granata I, Troiano E, Sangiovanni M, Guarracino MR. Integration of transcriptomic data in a genome-scale metabolic model to investigate the link between obesity and breast cancer. BMC Bioinformatics 2019; 20:162. [PMID: 30999849 PMCID: PMC6471692 DOI: 10.1186/s12859-019-2685-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Obesity is a complex disorder associated with an increased risk of developing several comorbid chronic diseases, including postmenopausal breast cancer. Although many studies have investigated this issue, the link between body weight and either risk or poor outcome of breast cancer is still to characterize. Systems biology approaches, based on the integration of multiscale models and data from a wide variety of sources, are particularly suitable for investigating the underlying molecular mechanisms of complex diseases. In this scenario, GEnome-scale metabolic Models (GEMs) are a valuable tool, since they represent the metabolic structure of cells and provide a functional scaffold for simulating and quantifying metabolic fluxes in living organisms through constraint-based mathematical methods. The integration of omics data into the structural information described by GEMs allows to build more accurate descriptions of metabolic states. RESULTS In this work, we exploited gene expression data of postmenopausal breast cancer obese and lean patients to simulate a curated GEM of the human adipocyte, available in the Human Metabolic Atlas database. To this aim, we used a published algorithm which exploits a data-driven approach to overcome the limitation of defining a single objective function to simulate the model. The flux solutions were used to build condition-specific graphs to visualise and investigate the reaction networks and their properties. In particular, we performed a network topology differential analysis to search for pattern differences and identify the principal reactions associated with significant changes across the two conditions under study. CONCLUSIONS Metabolic network models represent an important source to study the metabolic phenotype of an organism in different conditions. Here we demonstrate the importance of exploiting Next Generation Sequencing data to perform condition-specific GEM analyses. In particular, we show that the qualitative and quantitative assessment of metabolic fluxes modulated by gene expression data provides a valuable method for investigating the mechanisms associated with the phenotype under study, and can foster our interpretation of biological phenomena.
Collapse
Affiliation(s)
- Ilaria Granata
- High Performance Computing and Networking Institute, National Research Council of Italy, Via P. Castellino, 111, Napoli, 80131 Italy
| | - Enrico Troiano
- High Performance Computing and Networking Institute, National Research Council of Italy, Via P. Castellino, 111, Napoli, 80131 Italy
| | - Mara Sangiovanni
- Stazione Zoologica Anton Dohrn, Villa Comunale, Napoli, 80121 Italy
| | - Mario Rosario Guarracino
- High Performance Computing and Networking Institute, National Research Council of Italy, Via P. Castellino, 111, Napoli, 80131 Italy
| |
Collapse
|
19
|
Ayoub NM, Yaghan RJ, Abdo NM, Matalka II, Akhu-Zaheya LM, Al-Mohtaseb AH. Impact of Obesity on Clinicopathologic Characteristics and Disease Prognosis in Pre- and Postmenopausal Breast Cancer Patients: A Retrospective Institutional Study. J Obes 2019; 2019:3820759. [PMID: 31019808 PMCID: PMC6452538 DOI: 10.1155/2019/3820759] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/15/2019] [Accepted: 03/03/2019] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To investigate the association between obesity and breast cancer clinicopathologic characteristics at presentation along with prognostic impact among Jordanian breast cancer patients. Such data are lacking in Arabian countries. METHODS In this retrospective study, 348 breast cancer patients were included. Analyses were conducted for associations between body mass index (BMI) and age at diagnosis, tumor clinicopathologic characteristics, and molecular subtypes. Eight prognostic factors were considered, and total prognostic scores were calculated. The analysis was stratified by menopausal status. Multivariate logistic stepwise regression analysis was conducted to identify predictors for breast cancer recurrence and death. RESULTS Mean age at diagnosis was 50.98 ± 10.96 years. Mean BMI at diagnosis was 29.52 ± 5.32 kg/m2. Mean age at diagnosis was significantly higher for overweight and obese patients compared to underweight/normal patients (P < 0.001). A significant positive correlation was observed between patient age and BMI at diagnosis (r = 0.251, P < 0.001). Grade of carcinoma was significantly correlated with BMI in the whole population examined (P=0.003). Obese breast cancer patients had significantly higher prognostic scores compared to nonobese cases, indicating worse prognostic features at presentation (P=0.034). Stratification of data analysis based on menopausal status revealed significant associations between obesity and each of tumor stage and grade among postmenopausal but not premenopausal patients (P=0.019 and P=0.031, respectively). Similarly, postmenopausal obese patients had significantly higher prognostic scores compared to nonobese counterparts (P=0.007), indicating worse prognosis, a finding which was also absent among premenopausal breast cancer patients. No significant association between BMI with expression status of hormone receptors, HER2, lymphovascular invasion, and molecular subtypes was found among patients. BMI was a significant predictor for disease recurrence in which obese breast cancer patients had greater odds (2-fold) to develop locoregional and distant recurrence compared to nonobese cases (P=0.011). CONCLUSIONS Obesity was associated with advanced stage and grade of breast carcinoma at diagnosis. The impact of BMI on clinicopathologic characteristics and prognosis was confined to postmenopausal cases. Jordanian obese breast cancer patients are at greater risk of breast cancer recurrence and reduced survival compared to their nonobese counterparts.
Collapse
Affiliation(s)
- Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Rami J. Yaghan
- Department of Surgery, Faculty of Medicine, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Nour M. Abdo
- Department of Public Health and Community Medicine, Faculty of Medicine, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Ismail I. Matalka
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Laila M. Akhu-Zaheya
- Department of Adults Health Nursing, Faculty of Nursing, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Alia H. Al-Mohtaseb
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology (JUST), Irbid, Jordan
| |
Collapse
|
20
|
Molecular mechanisms linking high body mass index to breast cancer etiology in post-menopausal breast tumor and tumor-adjacent tissues. Breast Cancer Res Treat 2018; 173:667-677. [PMID: 30387004 DOI: 10.1007/s10549-018-5034-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/29/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE In post-menopausal women, high body mass index (BMI) is an established breast cancer risk factor and is associated with worse breast cancer prognosis. We assessed the associations between BMI and gene expression of both breast tumor and adjacent tissue in estrogen receptor-positive (ER+) and estrogen receptor-negative (ER-) diseases to help elucidate the mechanisms linking obesity with breast cancer biology in 519 post-menopausal women from the Nurses' Health Study (NHS) and NHSII. METHODS Differential gene expression was analyzed separately in ER+ and ER- disease both comparing overweight (BMI ≥ 25 to < 30) or obese (BMI ≥ 30) women to women with normal BMI (BMI < 25), and per 5 kg/m2 increase in BMI. Analyses controlled for age and year of diagnosis, physical activity, alcohol consumption, and hormone therapy use. Gene set enrichment analyses were performed and validated among a subset of post-menopausal cases in The Cancer Genome Atlas (for tumor) and Polish Breast Cancer Study (for tumor-adjacent). RESULTS No gene was differentially expressed by BMI (FDR < 0.05). BMI was significantly associated with increased cellular proliferation pathways, particularly in ER+ tumors, and increased inflammation pathways in ER- tumor and ER- tumor-adjacent tissues (FDR < 0.05). High BMI was associated with upregulation of genes involved in epithelial-mesenchymal transition in ER+ tumor-adjacent tissues. CONCLUSIONS This study provides insights into molecular mechanisms of BMI influencing post-menopausal breast cancer biology. Tumor and tumor-adjacent tissues provide independent information about potential mechanisms.
Collapse
|
21
|
Whole Genome Transcriptome Analysis of the Association between Obesity and Triple-Negative Breast Cancer in Caucasian Women. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:ijerph15112338. [PMID: 30360534 PMCID: PMC6265882 DOI: 10.3390/ijerph15112338] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/05/2018] [Accepted: 10/15/2018] [Indexed: 12/19/2022]
Abstract
Background: Triple-negative breast cancer (TNBC) is the most aggressive form of breast cancer, with poor outcomes. The molecular basis of TNBC remains poorly understood. The objective of this exploratory study was to investigate the association between obesity and TNBC in premenopausal and postmenopausal Caucasian women using transcription profiling. Methods: We compared gene expression levels of tumor samples drawn from normal weight, overweight, and obese pre and postmenopausal women diagnosed with TNBC. We performed hierarchical clustering to assess similarity in patterns of gene expression profiles, and conducted network and pathway analysis to identify molecular networks and biological pathways. Results: We discovered gene signatures distinguishing normal weight from obese, normal weight from overweight, and overweight from obese individuals in both premenopausal and postmenopausal women. The analysis revealed molecular networks and biological pathways associating obesity with TNBC. The discovered pathways included the unfolded protein response, endoplasmic reticulum stress, B cell receptor, and autophagy signaling pathways in obese premenopausal women; and the integrin, axonal guidance, ERK/MAPK (extracellular-signal-regulated kinase/mitogen activated protein kinase) and glutathione biosynthesis signaling pathways in obese postmenopausal women. Conclusions: The results suggest that both overweight and obese status are associated with TNBC, highlighting the need for conformation of these results in independent studies.
Collapse
|
22
|
Nuncia-Cantarero M, Martinez-Canales S, Andrés-Pretel F, Santpere G, Ocaña A, Galan-Moya EM. Functional transcriptomic annotation and protein-protein interaction network analysis identify NEK2, BIRC5, and TOP2A as potential targets in obese patients with luminal A breast cancer. Breast Cancer Res Treat 2018; 168:613-623. [PMID: 29330624 PMCID: PMC5842257 DOI: 10.1007/s10549-017-4652-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/29/2017] [Indexed: 12/22/2022]
Abstract
PURPOSE Although obesity is a risk factor for breast cancer, little effort has been made in the identification of druggable molecular alterations in obese-breast cancer patients. Tumors are controlled by their surrounding microenvironment, in which the adipose tissue is a main component. In this work, we intended to describe molecular alterations at a transcriptomic and protein-protein interaction (PPI) level between obese and non-obese patients. METHODS AND RESULTS Gene expression data of 269 primary breast tumors were compared between normal-weight (BMI < 25, n = 130) and obese (IMC > 30, n = 139) patients. No significant differences were found for the global breast cancer population. However, within the luminal A subtype, upregulation of 81 genes was observed in the obese group (FC ≥ 1.4). Next, we explored the association of these genes with patient outcome, observing that 39 were linked with detrimental outcome. Their PPI map formed highly compact cluster and functional annotation analyses showed that cell cycle, cell proliferation, cell differentiation, and cellular response to extracellular stimuli were the more altered functions. Combined analyses of genes within the described functions are correlated with poor outcome. PPI network analyses for each function were to search for druggable opportunities. We identified 16 potentially druggable candidates. Among them, NEK2, BIRC5, and TOP2A were also found to be amplified in breast cancer, suggesting that they could act as strategic players in the obese-deregulated transcriptome. CONCLUSION In summary, our in silico analysis describes molecular alterations of luminal A tumors and proposes a druggable PPI network in obese patients with potential for translation to the clinical practice.
Collapse
Affiliation(s)
- Miriam Nuncia-Cantarero
- Translational Oncology Laboratory, Centro Regional de Investigaciones Biomédicas (CRIB), Universidad de Castilla La Mancha (UCLM), C/Almansa 14, 02008, Albacete, Spain
| | | | | | - Gabriel Santpere
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Alberto Ocaña
- Translational Research Unit, University Hospital, Albacete, Spain
| | - Eva Maria Galan-Moya
- Translational Oncology Laboratory, Centro Regional de Investigaciones Biomédicas (CRIB), Universidad de Castilla La Mancha (UCLM), C/Almansa 14, 02008, Albacete, Spain.
| |
Collapse
|
23
|
Prieto D, Soto-Ferrari M, Tija R, Peña L, Burke L, Miller L, Berndt K, Hill B, Haghsenas J, Maltz E, White E, Atwood M, Norman E. Literature review of data-based models for identification of factors associated with racial disparities in breast cancer mortality. Health Syst (Basingstoke) 2018; 8:75-98. [PMID: 31275571 PMCID: PMC6598506 DOI: 10.1080/20476965.2018.1440925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 01/29/2018] [Accepted: 02/08/2018] [Indexed: 01/03/2023] Open
Abstract
In the United States, early detection methods have contributed to the reduction of overall breast cancer mortality but this pattern has not been observed uniformly across all racial groups. A vast body of research literature shows a set of health care, socio-economic, biological, physical, and behavioural factors influencing the mortality disparity. In this paper, we review the modelling frameworks, statistical tests, and databases used in understanding influential factors, and we discuss the factors documented in the modelling literature. Our findings suggest that disparities research relies on conventional modelling and statistical tools for quantitative analysis, and there exist opportunities to implement data-based modelling frameworks for (1) exploring mechanisms triggering disparities, (2) increasing the collection of behavioural data, and (3) monitoring factors associated with the mortality disparity across time.
Collapse
Affiliation(s)
- Diana Prieto
- College of Engineering and Applied Sciences, Western Michigan University, Kalamazoo, MI, USA
- Johns Hopkins Carey Business School, Baltimore, MD, USA
| | - Milton Soto-Ferrari
- College of Engineering and Applied Sciences, Western Michigan University, Kalamazoo, MI, USA
- Department of Marketing and Operations, Scott College of Business, Terre Haute, IN, USA
| | - Rindy Tija
- College of Engineering and Applied Sciences, Western Michigan University, Kalamazoo, MI, USA
| | - Lorena Peña
- College of Engineering and Applied Sciences, Western Michigan University, Kalamazoo, MI, USA
| | - Leandra Burke
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Lisa Miller
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Kelsey Berndt
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Brian Hill
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Jafar Haghsenas
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Ethan Maltz
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Evan White
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Maggie Atwood
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Earl Norman
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| |
Collapse
|
24
|
Song H, Lim DY, Jung JI, Cho HJ, Park SY, Kwon GT, Kang YH, Lee KW, Choi MS, Park JHY. Dietary oleuropein inhibits tumor angiogenesis and lymphangiogenesis in the B16F10 melanoma allograft model: a mechanism for the suppression of high-fat diet-induced solid tumor growth and lymph node metastasis. Oncotarget 2018; 8:32027-32042. [PMID: 28410190 PMCID: PMC5458266 DOI: 10.18632/oncotarget.16757] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 03/20/2017] [Indexed: 12/20/2022] Open
Abstract
Previously, we reported that high-fat-diet (HFD)-induced obesity stimulates melanoma progression in the B16F10 allograft model. In this study, we examined whether oleuropein (OL), the most abundant phenolic compound in olives, inhibits HFD-induced melanoma progression. Four-week-old male C57BL/6N mice were fed a HFD-diet with or without OL. After 16 weeks of feeding, B16F10-luc cells were subcutaneously injected and the primary tumor was resected 3 weeks later. OL suppressed HFD-induced solid tumor growth. In the tumor tissues, OL reduced HFD-induced expression of angiogenesis (CD31, VE-cadherin, VEGF-A, and VEGFR2), lymphangiogenesis (LYVE-1, VEGF-C, VEGF-D, and VEGFR3), and hypoxia (HIF-1α and GLUT-1) markers as well as HFD-induced increases in lipid vacuoles and M2 macrophages (MΦs). All animals were euthanized 2.5 weeks after tumor resection. OL suppressed HFD-induced increases in lymph node (LN) metastasis; expression of VEGF-A, VEGF-C, and VEGF-D in the LN; and M2-MΦs and the size of adipocytes in adipose tissues surrounding LNs. Co-culture results revealed that the crosstalk between B16F10s, M2-MΦs, and differentiated 3T3-L1 cells under hypoxic conditions increased the secretion of VEGF-A and -D, which stimulated tube formation and migration of endothelial cells (HUVECs) and lymphatic endothelial cells (LEC), respectively. Additionally, OL directly inhibited the differentiation of 3T3-L1 preadipocytes and tube formation by HUVECs and LECs. The overall results indicated that dietary OL inhibits lipid and M2-MΦ accumulation in HFD-fed mice, which contributes to decreases in VEGF secretion, thereby leading to inhibition of angiogenesis and lymphangiogenesis.
Collapse
Affiliation(s)
- Hyerim Song
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea
| | - Do Young Lim
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Jae In Jung
- Division of Bio-Imaging, Chuncheon Center, Korea Basic Science Institute, Chuncheon 24341, Republic of Korea
| | - Han Jin Cho
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea.,WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Republic of Korea
| | - So Young Park
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea.,Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea
| | - Gyoo Taik Kwon
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea.,Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea.,Berry and Biofood Research Institute, Jeonbuk 56417, Republic of Korea
| | - Young-Hee Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea
| | - Ki Won Lee
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Republic of Korea.,Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea.,Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Myung-Sook Choi
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jung Han Yoon Park
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea.,Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea.,Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
25
|
Cancer-related risk factors and incidence of major cancers by race, gender and region; analysis of the NIH-AARP diet and health study. BMC Cancer 2017; 17:597. [PMID: 28854891 PMCID: PMC5577755 DOI: 10.1186/s12885-017-3557-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 08/16/2017] [Indexed: 12/26/2022] Open
Abstract
Background Racial disparities in the incidence of major cancers may be attributed to differences in the prevalence of established, modifiable risk factors such as obesity, smoking, physical activity and diet. Methods Data from a prospective cohort of 566,398 adults aged 50–71 years, 19,677 African-American and 450,623 Whites, was analyzed. Baseline data on cancer-related risk factors such as smoking, alcohol, physical activity and dietary patterns were used to create an individual adherence score. Differences in adherence by race, gender and geographic region were assessed using descriptive statistics, and Cox proportional hazards models were used to determine the association between adherence and cancer incidence. Results Only 1.5% of study participants were adherent to all five cancer-related risk factor guidelines, with marked race-, gender- and regional differences in adherence overall. Compared with participants who were fully adherent to all five cancer risk factor criteria, those adherent to one or less had a 76% increased risk of any cancer incidence (HR: 1.76, 95% CI: 1.70 – 1.82), 38% increased risk of breast cancer (HR: 1.38, 95% CI: 1.25 – 1.52), and doubled the risk of colorectal cancer (HR: 2.06, 95% CI: 1.84 – 2.29). However, risk of prostate cancer was lower among participants adherent to one or less compared with those who were fully adherent (HR: 0.79, 95% CI: 0.75 – 0.85). The proportion of cancer incident cases attributable to low adherence was higher among African-Americans compared with Whites for all cancers (21% vs. 19%), and highest for colorectal cancer (25%) regardless of race. Conclusion Racial differences in the proportion of cancer incidence attributable to low adherence suggests unique opportunities for targeted cancer prevention strategies that may help eliminate racial disparities in cancer burden among older US adults. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3557-1) contains supplementary material, which is available to authorized users.
Collapse
|
26
|
Font-Clos F, Zapperi S, La Porta CAM. Integrative analysis of pathway deregulation in obesity. NPJ Syst Biol Appl 2017; 3:18. [PMID: 28685099 PMCID: PMC5493646 DOI: 10.1038/s41540-017-0018-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 05/22/2017] [Accepted: 05/31/2017] [Indexed: 12/14/2022] Open
Abstract
Obesity is a pandemic disease, linked to the onset of type 2 diabetes and cancer. Transcriptomic data provides a picture of the alterations in regulatory and metabolic activities associated with obesity, but its interpretation is typically blurred by noise. Here, we solve this problem by collecting publicly available transcriptomic data from adipocytes and removing batch effects using singular value decomposition. In this way we obtain a gene expression signature of 38 genes associated to obesity and identify the main pathways involved. We then show that similar deregulation patterns can be detected in peripheral markers, in type 2 diabetes and in breast cancer. The integration of different data sets combined with the study of pathway deregulation allows us to obtain a more complete picture of gene-expression patterns associated with obesity, breast cancer, and diabetes. The worldwide increase in obesity is extremely worrisome, especially because this condition is associated with a higher risk for diseases such as type 2 diabetes and cancer. Identifying alterations in regulatory and metabolic activities associated with obesity is complicated due to the presence of noise. A team lead by Caterina La Porta from the University of Milan addressed the question from the point of view of big data and extracted a signature of 38 genes associated to obesity from the combination of publicly available gene expression data from obese and lean subjects. The results revealed a similarity between the deregulation patterns observed in obesity and those found in breast cancer and diabetes, providing a clearer picture of the role of obesity in these diseases.
Collapse
Affiliation(s)
| | - Stefano Zapperi
- ISI Foundation, Via Chisola 5, 10126 Torino, Italy.,Center for Complexity and Biosystems, Department of Physics, Via Celoria 16, 20133 Milano, Italy.,CNR-Consiglio Nazionale delle Ricerche, Istituto di Chimica della Materia Condensata e di Tecnologie per l'Energia, Via R. Cozzi 53, 20125 Milano, Italy.,Department of Applied Physics, Aalto University, P.O. Box 11100, FIN-00076 Aalto, Finland
| | - Caterina A M La Porta
- Center for Complexity and Biosystems, Department of Environmental Science and Policy, University of Milano, via Celoria 26, 20133 Milano, Italy
| |
Collapse
|
27
|
Zhao YX, Liu S, Hu YB, Ge YY, Lv DM. Diagnostic and prognostic values of contrast-enhanced ultrasound in breast cancer: a retrospective study. Onco Targets Ther 2017; 10:1123-1129. [PMID: 28260926 PMCID: PMC5328613 DOI: 10.2147/ott.s124134] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This study aimed to explore the diagnostic and prognostic values of contrast-enhanced ultrasound (CEUS) in breast cancer. Between September 2009 and October 2011, a total of 143 breast cancer patients and 161 healthy people were selected as case group and control group, respectively. After the identification of lesions by conventional ultrasound, all patients underwent CEUS. The CEUS images were analyzed, and time-intensity curves (TICs) were obtained. Hematoxylin-eosin and immunohistochemistry staining was performed on tissue specimens, according to which the expressions of estrogen receptor (ER), c-erb-B2, p53, and Ki-67 were measured. Multivariate logistic regression analysis was used to compare CEUS and TIC parameters between the two groups. Compared with the control group, cancer patients showed high enhancement, heterogeneous enhancement or defects in the central region, expansion of lesion diameter after enhancement and crab-like blur lesion edges. The peak intensity (PI), relative start time of enhancement, relative PI, and relative area under the curve in the case group were significantly higher than those in the control group. Logistic analysis showed that the uniformity of enhancement, expansion of lesion diameter, and relative PI were significant diagnostic parameters of breast cancer, with area under the curve being 0.798, 0.776, and 0.919, respectively. There were strong associations between CEUS characteristics and expressions of prognostic factors in breast cancer: the heterogeneous enhancement was common in c-erb-B2-positive tumors; the centripetal enhancement occurred more in ER-negative tumors; perforator vessels were often seen in tumors at high histological grade; perfusion defects were common in ER-negative, c-erb-B2-positive, and Ki-67-positive tumors. CEUS is a useful tool for the early diagnosis and prognosis of breast cancer.
Collapse
Affiliation(s)
- Yi-Xuan Zhao
- Department of Ultrasound, Second Hospital, Jilin University, Changchun, People’s Republic of China
| | - Shuang Liu
- Department of Ultrasound, Second Hospital, Jilin University, Changchun, People’s Republic of China
| | - Yan-Bing Hu
- Department of Ultrasound, Second Hospital, Jilin University, Changchun, People’s Republic of China
| | - Yan-Yan Ge
- Department of Ultrasound, Second Hospital, Jilin University, Changchun, People’s Republic of China
| | - Dong-Mei Lv
- Department of Ultrasound, Second Hospital, Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
28
|
Abstract
Obesity rates are increasing in the developed and developing world; this has implications for breast cancer risk and outcome. Areas covered: Recent advances relating to the association of obesity with breast cancer are reviewed. Expert commentary: Obesity has been associated with increased risk of postmenopausal hormone receptor positive and premenopausal triple negative breast cancer and with poor prognosis of most types of breast cancer. Obese individuals may present with breast cancer at a more advanced stage and their breast cancer may differ biologically from cancers diagnosed in nonobese women. A picture of a complex, multifactorial biology underlying the obesity-cancer link is emerging, with the identification of obesity-associated tissue and systemic changes that are cancer promoting, enhancing proliferation, invasion and metastasis. Intervention research to ascertain effects of weight loss and of pharmacologic interventions that reverse the metabolic changes of obesity is needed.
Collapse
Affiliation(s)
- Pamela J Goodwin
- a Department of Medicine, Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital, University of Toronto , Toronto , Canada
| |
Collapse
|