1
|
Kaushal JB, Raut P, Muniyan S, Siddiqui JA, Alsafwani ZW, Seshacharyulu P, Nair SS, Tewari AK, Batra SK. Racial disparity in prostate cancer: an outlook in genetic and molecular landscape. Cancer Metastasis Rev 2024; 43:1233-1255. [PMID: 38902476 PMCID: PMC11560487 DOI: 10.1007/s10555-024-10193-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/04/2024] [Indexed: 06/22/2024]
Abstract
Prostate cancer (PCa) incidence, morbidity, and mortality rates are significantly impacted by racial disparities. Despite innovative therapeutic approaches and advancements in prevention, men of African American (AA) ancestry are at a higher risk of developing PCa and have a more aggressive and metastatic form of the disease at the time of initial PCa diagnosis than other races. Research on PCa has underlined the biological and molecular basis of racial disparity and emphasized the genetic aspect as the fundamental component of racial inequality. Furthermore, the lower enrollment rate, limited access to national-level cancer facilities, and deferred treatment of AA men and other minorities are hurdles in improving the outcomes of PCa patients. This review provides the most up-to-date information on various biological and molecular contributing factors, such as the single nucleotide polymorphisms (SNPs), mutational spectrum, altered chromosomal loci, differential gene expression, transcriptome analysis, epigenetic factors, tumor microenvironment (TME), and immune modulation of PCa racial disparities. This review also highlights future research avenues to explore the underlying biological factors contributing to PCa disparities, particularly in men of African ancestry.
Collapse
Affiliation(s)
- Jyoti B Kaushal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Pratima Raut
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Sakthivel Muniyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Jawed A Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Zahraa W Alsafwani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Parthasarathy Seshacharyulu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Sujit S Nair
- Department of Urology and the Tisch Cancer Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ashutosh K Tewari
- Department of Urology and the Tisch Cancer Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA.
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE-68198, USA.
- Division of Urology, Department of Surgery, University of Nebraska Medical Center, Omaha, NE-68198, USA.
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE-68198, USA.
| |
Collapse
|
2
|
Tseng JC, Wang BJ, Wang YP, Kuo YY, Chen JK, Hour TC, Kuo LK, Hsiao PJ, Yeh CC, Kao CL, Shih LJ, Chuu CP. Caffeic acid phenethyl ester suppresses EGFR/FAK/Akt signaling, migration, and tumor growth of prostate cancer cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154860. [PMID: 37201366 DOI: 10.1016/j.phymed.2023.154860] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) is upregulated in prostate cancer (PCa). However, suppression of EGFR did not improve the patient outcome, possibly due to the activation of PI3K/Akt signaling in PCa. Compounds able to suppress both PI3K/Akt and EGFR signaling may be effective for treating advanced PCa. PURPOSE We examined if caffeic acid phenethyl ester (CAPE) simultaneously suppresses the EGFR and Akt signaling, migration and tumor growth in PCa cells. METHODS Wound healing assay, transwell migration assay and xenograft mice model were used to determine the effects of CAPE on migration and proliferation of PCa cells. Western blot, immunoprecipitation, and immunohistochemistry staining were performed to determine the effects of CAPE on EGFR and Akt signaling. RESULTS CAPE treatment decreased the gene expression of HRAS, RAF1, AKT2, GSK3A, and EGF and the protein expression of phospho-EGFR (Y845, Y1069, Y1148, Y1173), phospho-FAK, Akt, and ERK1/2 in PCa cells. CAPE treatment inhibited the EGF-induced migration of PCa cells. Combined treatment of CAPE with EGFR inhibitor gefitinib showed additive inhibition on migration and proliferation of PCa cells. Injection of CAPE (15 mg/kg/3 days) for 14 days suppressed the tumor growth of prostate xenografts in nude mice as well as suppressed the levels of Ki67, phospho-EGFR Y845, MMP-9, phospho-Akt S473, phospho-Akt T308, Ras, and Raf-1 in prostate xenografts. CONCLUSIONS Our study suggested that CAPE can simultaneously suppress the EGFR and Akt signaling in PCa cells and is a potential therapeutic agent for advanced PCa.
Collapse
Affiliation(s)
- Jen-Chih Tseng
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County 35053, Taiwan; Immunology Research Center, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Bi-Juan Wang
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Ya-Pei Wang
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Ying-Yu Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Jen-Kun Chen
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Tzyh-Chyuan Hour
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung 80737, Taiwan; Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung 80737, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80737, Taiwan
| | - Li-Kuo Kuo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mackay Memorial Hospital, Taipei City 104217, Taiwan; Department of Nursing, Mackay Medical College, Taipei City, Taiwan
| | - Po-Jen Hsiao
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County 35053, Taiwan; Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan 325, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chien-Chih Yeh
- Department of Education and Medical Research, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Colon and Rectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Li Kao
- Division of Urology, Departments of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Division of Urology, Department of Surgery, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Li-Jane Shih
- Department of Education and Medical Research, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Graduate Institute of Medical Science, National Defense Medical Center, Taipei 114, Taiwan
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County 35053, Taiwan; Department of Life Sciences, National Central University, Taoyuan 32031, Taiwan; PhD Program for Aging and Graduate Institute of Basic Medical Science, China Medical University, Taichung City 40402, Taiwan; Biotechnology Center, National Chung Hsing University, Taichung City 40227, Taiwan.
| |
Collapse
|
3
|
Zhang B, Yao K, Cheng C. A network-based integration for understanding racial disparity in prostate cancer. Transl Oncol 2022; 17:101327. [PMID: 34998235 PMCID: PMC8738961 DOI: 10.1016/j.tranon.2021.101327] [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] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 11/24/2022] Open
Abstract
Compared to Caucasians (CAs), African Americans (AAs) have a higher rate of incidence and mortality in prostate cancer and are prone to be diagnosed at later stages. To understand this racial disparity, molecular features of different types, including gene expression, DNA methylation and other genomic alterations, have been compared between tumor samples from the two races, but led to different disparity associated genes (DAGs). In this study, we applied a network-based algorithm to integrate a comprehensive set of genomic datasets and identified 130 core DAGs. Out of these genes, 78 were not identified by any individual dataset but prioritized and selected through network propagation. We found DAGs were highly enriched in several critical prostate cancer-related signaling transduction and cell cycle pathways and were more likely to be associated with patient prognosis in prostate cancer. Furthermore, DAGs were over-represented in prostate cancer risk genes identified from previous genome wide association studies. We also found DAGs were enriched in kinase and transcription factor encoding genes. Interestingly, for many of these prioritized kinases their association with racial disparity did not manifest from the original genomic/transcriptomic data but was reflected by their differential phosphorylation levels between AA and CA prostate tumor samples. Similarly, the disparity relevance of some transcription factors was not reflected at the mRNA or protein expression level, but at the activity level as demonstrated by their differential ability in regulating target gene expression. Our integrative analysis provided new candidate targets for improving prostate cancer treatment and addressing the racial disparity problem.
Collapse
Affiliation(s)
- Baoyi Zhang
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX 77030, United States
| | - Kevin Yao
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, United States
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, United States; Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, United States; Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX 77030, United States.
| |
Collapse
|
4
|
Abraham-Miranda J, Awasthi S, Yamoah K. Immunologic disparities in prostate cancer between American men of African and European descent. Crit Rev Oncol Hematol 2021; 164:103426. [PMID: 34273500 DOI: 10.1016/j.critrevonc.2021.103426] [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: 07/08/2020] [Revised: 12/18/2020] [Accepted: 07/12/2021] [Indexed: 11/27/2022] Open
Abstract
Health disparities between American men of African and European descent (AA and EA, respectively) can be attributed to multiple factors, including disparities in socioeconomic status, access to healthcare, lifestyle, ancestry, and molecular aberrations. Numerous clinical trials and research studies are being performed to identify new and better therapeutic approaches to detect and treat prostate cancer. Of potential concern is the fact that the majority of the patients enrolled on these trials are EA. This disproportionate enrollment of EA could have implications when disease management recommendations are proposed without regard to the existing disparities in prostate cancer between races. With increasing advancements in immunotherapies, the immunological disparities between men of diverse ethnicities will need to be fully explored to develop novel and effective therapeutic approaches for prostate cancer patients globally. To help address this need, this review fully describes inequalities in prostate cancer at the immunological level between AA and EA.
Collapse
Affiliation(s)
- Julieta Abraham-Miranda
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Shivanshu Awasthi
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Kosj Yamoah
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA; Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
5
|
Mishra S, Charan M, Verma AK, Ramaswamy B, Ahirwar DK, Ganju RK. Racially Disparate Expression of mTOR/ERK-1/2 Allied Proteins in Cancer. Front Cell Dev Biol 2021; 9:601929. [PMID: 33996789 PMCID: PMC8120233 DOI: 10.3389/fcell.2021.601929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Recent studies revealed that ethnic differences in mechanistic target of rapamycin (mTOR) and extracellular signal-regulated kinase (ERK-1/2) signaling pathways might be associated with the development and progression of different human malignancies. The African American (AA) population has an increased rate of cancer incidence and mortality compared to the Caucasian American (CA) population. Although the socioeconomic differences across different ethnic groups contribute to the disparity in developing different cancers, recent scientific evidence indicates the association of molecular and genetic variations in racial disparities of different human malignancies. The mTOR and ERK-1/2 signaling pathways are one of the well-known oncogenic signaling mechanisms that regulate diverse molecular and phenotypic aspects of normal as well as cancer cells in response to different external or internal stimuli. To date, very few studies have been carried out to explore the significance of racial disparity with abnormal mTOR and ERK-1/2 kinase signaling pathways, which may contribute to the development of aggressive human cancers. In this review, we discuss the differential regulation of mTOR and ERK-1/2 kinase signaling pathways across different ethnic groups, especially between AA and CA populations. Notably, we observed that key signaling proteins associated with mTOR and ERK-1/2 pathway including transforming growth factor-beta (TGF-β), Akt, and VEGFR showed racially disparate expression in cancer patients. Overall, this review article encompasses the significance of racially disparate signaling molecules related to mTOR/ERK1/2 and their potential in developing tailor-made anti-cancer therapies.
Collapse
Affiliation(s)
- Sanjay Mishra
- Department of Pathology, Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Manish Charan
- Department of Pathology, Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Ajeet Kumar Verma
- Department of Pathology, Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, OH, United States
| | | | - Dinesh Kumar Ahirwar
- Department of Pathology, Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Ramesh K Ganju
- Department of Pathology, Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, OH, United States.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
6
|
Bahcivan A, Gamsizkan M, Kantarcioglu Coskun S, Cangur S, Yuksel A, Ceyhan A, Onal B. KRAS, BRAF, PIK3CA mutation frequency of radical prostatectomy samples and review of the literature. Aging Male 2020; 23:1627-1641. [PMID: 33878842 DOI: 10.1080/13685538.2021.1901274] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE The molecular basis of prostate cancer is highly heterogeneous. Our study aimed to perform the mutation analysis of KRAS, BRAF, PIK3CA, and immunohistochemical (IHC) evaluation of EGFR, HER2, p16, and PTEN to demonstrate new areas for targeted therapies. METHODS A total of 24 prostatectomy samples diagnosed with adenocarcinoma were analyzed by microarray hybridization. Also, these samples were IHC stained for EGFR, HER2, P16, and PTEN. The cases were divided into two groups based on low and high Gleason scores. All findings were compared with the clinicopathological parameters of the patients. RESULTS While KRAS mutation was in 3/24 (12.5%) of our cases, BRAF and PIK3CA mutations were not detected. There was no significant difference between the groups in terms of KRAS mutation frequency. HER2 was immunohistochemically negative in all samples. There was no correlation between EGFR, P16 immunopositivity, and clinicopathological features. CONCLUSION KRAS mutation frequency is similar to those in Asian populations. BRAF and PIK3CA mutation frequencies have been reported in the literature in the range of 0-15% and 0-10.4%, respectively, consistent with our study findings. HER2 immunoexpression is a controversial issue in the literature. EGFR and p16 expressions may not correlate with the stage.
Collapse
Affiliation(s)
- Atike Bahcivan
- Department of Pathology, Duzce University, Duzce, Turkey
| | | | | | - Sengul Cangur
- Department of Biostatistics and Medical Informatics, Duzce University, Duzce, Turkey
| | | | - Aysegul Ceyhan
- Department of Pathology, Duzce University, Duzce, Turkey
| | - Binnur Onal
- Department of Pathology, Duzce University, Duzce, Turkey
| |
Collapse
|
7
|
Poulson MR, Helrich SA, Kenzik KM, Dechert TA, Sachs TE, Katz MH. The impact of racial residential segregation on prostate cancer diagnosis and treatment. BJU Int 2020; 127:636-644. [PMID: 33166036 DOI: 10.1111/bju.15293] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES To examine the effects of racial residential segregation and structural racism on the diagnosis, treatment, and outcomes of patients with prostate cancer. PATIENTS AND METHODS This retrospective cohort study examined men diagnosed with prostate cancer between 2005 and 2015. We collected data from Black and White men, aged ≥30 years, living within the 100 most populous counties participating in the Surveillance, Epidemiology, and End Results programme, a nationally representative dataset. The racial Index of Dissimilarity, a validated measure of segregation, was the primary exposure of interest. Outcomes of interest included advanced stage at diagnosis (Stage IV), surgery for localised disease (Stage I-II), and 10-year overall and cancer-specific survival. Multivariable Poisson regression analyses with robust error variance estimated the relative risk (RR) of advanced stage at diagnosis and surgery for localised disease at differing levels of segregation. Survival analysis was performed using competing hazards analysis. RESULTS Multivariable models estimating stage at diagnosis showed that the disparities between Black and White men disappeared at low levels of segregation. Disparities in receiving surgery for localised disease persisted across all levels of segregation. In racially stratified analyses, segregation had no effect on stage at diagnosis or surgical resection for Black patients. White patients saw a 56% (RR 0.42, P < 0.001) reduced risk of presenting at advanced stage and 20% increased likelihood (RR 1.20, P < 0.001) of surgery for localised disease. Black patients in the lowest segregation areas had the lowest overall mortality, but the highest cancer-specific mortality. CONCLUSIONS Our study provides evidence that residential segregation has a significant impact on Black-White disparities in prostate cancer, likely through improved outcomes for White patients and worse outcomes for Black patients in more segregated areas. These findings suggest that mitigating segregation and the downstream effects of socioeconomic factors could alleviate these disparities.
Collapse
Affiliation(s)
- Michael R Poulson
- Department of Surgery, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Samuel A Helrich
- Department of Urology, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Kelly M Kenzik
- Department of Surgery, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA.,Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tracey A Dechert
- Department of Surgery, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Teviah E Sachs
- Department of Surgery, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Mark H Katz
- Department of Urology, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
8
|
Davis M, Martini R, Newman L, Elemento O, White J, Verma A, Datta I, Adrianto I, Chen Y, Gardner K, Kim HG, Colomb WD, Eltoum IE, Frost AR, Grizzle WE, Sboner A, Manne U, Yates C. Identification of Distinct Heterogenic Subtypes and Molecular Signatures Associated with African Ancestry in Triple Negative Breast Cancer Using Quantified Genetic Ancestry Models in Admixed Race Populations. Cancers (Basel) 2020; 12:E1220. [PMID: 32414099 PMCID: PMC7281131 DOI: 10.3390/cancers12051220] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
Triple negative breast cancers (TNBCs) are molecularly heterogeneous, and the link between their aggressiveness with African ancestry is not established. We investigated primary TNBCs for gene expression among self-reported race (SRR) groups of African American (AA, n = 42) and European American (EA, n = 33) women. RNA sequencing data were analyzed to measure changes in genome-wide expression, and we utilized logistic regressions to identify ancestry-associated gene expression signatures. Using SNVs identified from our RNA sequencing data, global ancestry was estimated. We identified 156 African ancestry-associated genes and found that, compared to SRR, quantitative genetic analysis was a more robust method to identify racial/ethnic-specific genes that were differentially expressed. A subset of African ancestry-specific genes that were upregulated in TNBCs of our AA patients were validated in TCGA data. In AA patients, there was a higher incidence of basal-like two tumors and altered TP53, NFB1, and AKT pathways. The distinct distribution of TNBC subtypes and altered oncologic pathways show that the ethnic variations in TNBCs are driven by shared genetic ancestry. Thus, to appreciate the molecular diversity of TNBCs, tumors from patients of various ancestral origins should be evaluated.
Collapse
Affiliation(s)
- Melissa Davis
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA; (M.D.); (R.M.); (L.N.)
| | - Rachel Martini
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA; (M.D.); (R.M.); (L.N.)
| | - Lisa Newman
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA; (M.D.); (R.M.); (L.N.)
| | - Olivier Elemento
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA;
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA;
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jason White
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, AL 36088, USA; (J.W.); (W.D.C.)
| | - Akanksha Verma
- Department of Computational Biology, Weill Cornell Medicine, New York, NY 10065, USA;
| | - Indrani Datta
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI 48202, USA; (I.D.); (I.A.); (Y.C.)
| | - Indra Adrianto
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI 48202, USA; (I.D.); (I.A.); (Y.C.)
| | - Yalei Chen
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI 48202, USA; (I.D.); (I.A.); (Y.C.)
| | - Kevin Gardner
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10027, USA;
| | - Hyung-Gyoon Kim
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (H.-G.K.); (I.-E.E.); (A.R.F.); (W.E.G.)
| | - Windy D. Colomb
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, AL 36088, USA; (J.W.); (W.D.C.)
- Department of Hematology and Oncology, Our Lady of Lourdes JD Moncus Cancer Center, Lafayette, LA 70508, USA
| | - Isam-Eldin Eltoum
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (H.-G.K.); (I.-E.E.); (A.R.F.); (W.E.G.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Andra R. Frost
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (H.-G.K.); (I.-E.E.); (A.R.F.); (W.E.G.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - William E. Grizzle
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (H.-G.K.); (I.-E.E.); (A.R.F.); (W.E.G.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Andrea Sboner
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA;
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10062, USA
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (H.-G.K.); (I.-E.E.); (A.R.F.); (W.E.G.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Clayton Yates
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, AL 36088, USA; (J.W.); (W.D.C.)
| |
Collapse
|
9
|
Das H, Rodriguez R. Health Care Disparities in Urologic Oncology: A Systematic Review. Urology 2020; 136:9-18. [DOI: 10.1016/j.urology.2019.09.058] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/12/2019] [Accepted: 09/23/2019] [Indexed: 12/21/2022]
|
10
|
|
11
|
Grant DJ, Manichaikul A, Alberg AJ, Bandera EV, Barnholtz‐Sloan J, Bondy M, Cote ML, Funkhouser E, Moorman PG, Peres LC, Peters ES, Schwartz AG, Terry PD, Wang X, Keku TO, Hoyo C, Berchuck A, Sandler DP, Taylor JA, O’Brien KM, Velez Edwards DR, Edwards TL, Beeghly‐Fadiel A, Wentzensen N, Pearce CL, Wu AH, Whittemore AS, McGuire V, Sieh W, Rothstein JH, Modugno F, Ness R, Moysich K, Rossing MA, Doherty JA, Sellers TA, Permuth‐Way JB, Monteiro AN, Levine DA, Setiawan VW, Haiman CA, LeMarchand L, Wilkens LR, Karlan BY, Menon U, Ramus S, Gayther S, Gentry‐Maharaj A, Terry KL, Cramer DW, Goode EL, Larson MC, Kaufmann SH, Cannioto R, Odunsi K, Etter JL, Huang R, Bernardini MQ, Tone AA, May T, Goodman MT, Thompson PJ, Carney ME, Tworoger SS, Poole EM, Lambrechts D, Vergote I, Vanderstichele A, Van Nieuwenhuysen E, Anton‐Culver H, Ziogas A, Brenton JD, Bjorge L, Salvensen HB, Kiemeney LA, Massuger LFAG, Pejovic T, Bruegl A, Moffitt M, Cook L, Le ND, Brooks‐Wilson A, Kelemen LE, Pharoah PD, Song H, Campbell I, Eccles D, DeFazio A, Kennedy CJ, Schildkraut JM. Evaluation of vitamin D biosynthesis and pathway target genes reveals UGT2A1/2 and EGFR polymorphisms associated with epithelial ovarian cancer in African American Women. Cancer Med 2019; 8:2503-2513. [PMID: 31001917 PMCID: PMC6536963 DOI: 10.1002/cam4.1996] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 12/03/2018] [Accepted: 01/08/2019] [Indexed: 02/02/2023] Open
Abstract
An association between genetic variants in the vitamin D receptor (VDR) gene and epithelial ovarian cancer (EOC) was previously reported in women of African ancestry (AA). We sought to examine associations between genetic variants in VDR and additional genes from vitamin D biosynthesis and pathway targets (EGFR, UGT1A, UGT2A1/2, UGT2B, CYP3A4/5, CYP2R1, CYP27B1, CYP24A1, CYP11A1, and GC). Genotyping was performed using the custom-designed 533,631 SNP Illumina OncoArray with imputation to the 1,000 Genomes Phase 3 v5 reference set in 755 EOC cases, including 537 high-grade serous (HGSOC), and 1,235 controls. All subjects are of African ancestry (AA). Logistic regression was performed to estimate odds ratios (OR) and 95% confidence intervals (CI). We further evaluated statistical significance of selected SNPs using the Bayesian False Discovery Probability (BFDP). A significant association with EOC was identified in the UGT2A1/2 region for the SNP rs10017134 (per allele OR = 1.4, 95% CI = 1.2-1.7, P = 1.2 × 10-6 , BFDP = 0.02); and an association with HGSOC was identified in the EGFR region for the SNP rs114972508 (per allele OR = 2.3, 95% CI = 1.6-3.4, P = 1.6 × 10-5 , BFDP = 0.29) and in the UGT2A1/2 region again for rs1017134 (per allele OR = 1.4, 95% CI = 1.2-1.7, P = 2.3 × 10-5 , BFDP = 0.23). Genetic variants in the EGFR and UGT2A1/2 may increase susceptibility of EOC in AA women. Future studies to validate these findings are warranted. Alterations in EGFR and UGT2A1/2 could perturb enzyme efficacy, proliferation in ovaries, impact and mark susceptibility to EOC.
Collapse
Affiliation(s)
- Delores J. Grant
- Department of Biological and Biomedical Sciences, Cancer Research ProgramJLC‐Biomedical/Biotechnology Research Institute, North Carolina Central UniversityDurhamNorth Carolina
| | - Ani Manichaikul
- Center for Public Health GenomicsUniversity of VirginiaCharlottesvilleVirginia
| | - Anthony J. Alberg
- Department of Epidemiology and Biostatistics, Arnold School of Public HealthUniversity of South CarolinaColumbiaSouth Carolina
| | - Elisa V. Bandera
- Department of Population ScienceRutgers Cancer Institute of New JerseyNew BrunswickNew Jersey
| | - Jill Barnholtz‐Sloan
- Case Comprehensive Cancer CenterCase Western Reserve University School of MedicineClevelandOhio
| | - Melissa Bondy
- Cancer Prevention and Population Sciences ProgramBaylor College of MedicineHoustonTexas
| | - Michele L. Cote
- Department of Oncology and the Karmanos Cancer Institute Population Studies and Disparities Research ProgramWayne State University School of MedicineDetroitMichigan
| | - Ellen Funkhouser
- Division of Preventive MedicineUniversity of Alabama at BirminghamBirminghamAlabama
| | - Patricia G. Moorman
- Department of Community and Family MedicineDuke University Medical CenterDurhamNorth Carolina
| | - Lauren C. Peres
- Center for Public Health GenomicsUniversity of VirginiaCharlottesvilleVirginia
| | - Edward S. Peters
- Epidemiology ProgramLouisiana State University Health Sciences Center School of Public HealthNew OrleansLouisisana
| | - Ann G. Schwartz
- Department of Oncology and the Karmanos Cancer Institute Population Studies and Disparities Research ProgramWayne State University School of MedicineDetroitMichigan
| | - Paul D. Terry
- Department of MedicineUniversity of Tennessee Medical Center – KnoxvilleKnoxvilleTennessee
| | - Xin‐Qun Wang
- Department of Public Health SciencesUniversity of VirginiaCharlottesvilleVirginia
| | - Temitope O. Keku
- Departments of Medicine and Nutrition, Division of Gastroenterology and HepatologyUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Cathrine Hoyo
- Department of Biological SciencesNorth Carolina State UniversityRaleighNorth Carolina
| | - Andrew Berchuck
- Department of Obstetrics and GynecologyDuke University Medical CenterDurhamNorth Carolina
| | - Dale P. Sandler
- Epidemiology Branch, Division of Intramural ResearchNational Institute of Environmental Health Sciences, National Institutes of HealthResearch Triangle ParkNorth Carolina
| | - Jack A. Taylor
- Epidemiology Branch, Division of Intramural ResearchNational Institute of Environmental Health Sciences, National Institutes of HealthResearch Triangle ParkNorth Carolina
| | - Katie M. O’Brien
- Epidemiology Branch, Division of Intramural ResearchNational Institute of Environmental Health Sciences, National Institutes of HealthResearch Triangle ParkNorth Carolina
| | - Digna R. Velez Edwards
- Vanderbilt Epidemiology Center, Center for Human Genetics Research, Department of Obstetrics and GynecologyVanderbilt University Medical CenterNashvilleTennessee
| | - Todd L. Edwards
- Division of Epidemiology, Center for Human Genetics Research, Department of MedicineVanderbilt University Medical CenterNashvilleTennessee
| | - Alicia Beeghly‐Fadiel
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology CenterInstitute for Medicine and Public Health, Vanderbilt University Medical CenterNashvilleTennessee
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMaryland
| | - Celeste Leigh Pearce
- Department of EpidemiologyUniversity of Michigan School of Public HealthAnn ArborMichigan
- Department of Preventive Medicine, Keck School of MedicineUniversity of Southern California Norris Comprehensive Cancer CenterLos AngelesCalifornia
| | - Anna H. Wu
- Department of Preventive Medicine, Keck School of MedicineUniversity of Southern California Norris Comprehensive Cancer CenterLos AngelesCalifornia
| | - Alice S. Whittemore
- Department of Health Research and PolicyStanford University School of MedicineStanfordCalifornia
- Department of Biomedical Data ScienceStanford University School of MedicineStanfordCalifornia
| | - Valerie McGuire
- Department of Health Research and PolicyStanford University School of MedicineStanfordCalifornia
| | - Weiva Sieh
- Department of Population Health Science and PolicyIcahn School of Medicine at Mount SinaiNew YorkNew York
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew York
| | - Joseph H. Rothstein
- Department of Population Health Science and PolicyIcahn School of Medicine at Mount SinaiNew YorkNew York
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew York
| | - Francesmary Modugno
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive SciencesUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
- Department of EpidemiologyUniversity of Pittsburgh Graduate School of Public HealthPittsburghPennsylvania
- Ovarian Cancer Center of Excellence, Womens Cancer Research ProgramMagee‐Womens Research Institute and University of Pittsburgh Cancer InstitutePittsburghPennsylvania
| | - Roberta Ness
- The University of Texas School of Public HealthHoustonTexas
| | - Kirsten Moysich
- Department of Cancer Prevention and ControlRoswell Park Cancer InstituteBuffaloNew York
| | - Mary Anne Rossing
- Program in Epidemiology, Division of Public Health SciencesFred Hutchinson Cancer Research CenterSeattleWashington
- Department of EpidemiologyUniversity of WashingtonSeattleWashington
| | - Jennifer A. Doherty
- Department of Population Health SciencesHuntsman Cancer Institute, University of UtahSalt Lake City, Utah
| | | | | | | | - Douglas A. Levine
- Gynecology Service, Department of SurgeryMemorial Sloan Kettering Cancer CenterNew YorkNew York
- Gynecologic Oncology, Laura and Isaac Pearlmutter Cancer CenterNew York University Langone Medical CenterNew YorkNew York
| | | | - Christopher A. Haiman
- University of Southern California Norris Comprehensive Cancer CenterLos AngelesCalifornia
| | | | - Lynne R. Wilkens
- Cancer Epidemiology ProgramUniversity of Hawaii Cancer CenterHawaii
| | - Beth Y. Karlan
- Women's Cancer ProgramSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCalifornia
| | - Usha Menon
- MRC CTU at UCL, Institute of Clinical Trials and MethodologyUniversity College LondonLondonUK
| | - Susan Ramus
- School of Women's and Children's HealthUniversity of New South WalesNew South WalesAustralia
- The Kinghorn Cancer CentreGarvan Institute of Medical ResearchDarlinghurstNew South WalesAustralia
| | - Simon Gayther
- Center for Cancer Prevention and Translational GenomicsSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCalifornia
- Department of Biomedical SciencesCedars‐Sinai Medical CenterLos AngelesCalifornia
| | | | - Kathryn L. Terry
- Obstetrics and Gynecology Epidemiology CenterBrigham and Women's HospitalBostonMassachusetts
- Harvard T. H. Chan School of Public HealthBostonMassauchusetts
| | - Daniel W. Cramer
- Obstetrics and Gynecology Epidemiology CenterBrigham and Women's HospitalBostonMassachusetts
- Harvard T. H. Chan School of Public HealthBostonMassauchusetts
| | - Ellen L. Goode
- Department of Health Science Research, Division of EpidemiologyMayo ClinicRochesterMinnesota
| | - Melissa C. Larson
- Department of Health Science Research, Division of Biomedical Statistics and InformaticsMayo ClinicRochesterMinnesota
| | - Scott H. Kaufmann
- Departments of Medicine and PharmacologyMayo ClinicRochesterMinnesota
| | - Rikki Cannioto
- Cancer Pathology & Prevention, Division of Cancer Prevention and Population SciencesRoswell Park Cancer InstituteBuffaloNew York
| | - Kunle Odunsi
- Department of Gynecological OncologyRoswell Park Cancer InstituteBuffaloNew York
| | - John L. Etter
- Department of Cancer Prevention and ControlRoswell Park Cancer InstituteBuffaloNew York
| | - Ruea‐Yea Huang
- Center For ImmunotherapyRoswell Park Cancer InstituteBuffaloNew York
| | - Marcus Q. Bernardini
- Division of Gynecologic OncologyPrincess Margaret Hospital, University Health NetworkTorontoOntarioCanada
| | - Alicia A. Tone
- Division of Gynecologic OncologyPrincess Margaret Hospital, University Health NetworkTorontoOntarioCanada
| | - Taymaa May
- Division of Gynecologic OncologyPrincess Margaret Hospital, University Health NetworkTorontoOntarioCanada
| | - Marc T. Goodman
- Cancer Prevention and ControlSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCalifornia
- Department of Biomedical SciencesCommunity and Population Health Research Institute, Cedars‐Sinai Medical CenterLos AngelesCalifornia
| | - Pamela J. Thompson
- Cancer Prevention and ControlSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCalifornia
| | - Michael E. Carney
- Department of Obstetrics and GynecologyJohn A. Burns School of Medicine, University of HawaiiHonoluluHawaii
| | - Shelley S. Tworoger
- Channing Division of Network MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusetts
| | | | - Diether Lambrechts
- Vesalius Research Center, VIBLeuvenBelgium
- Laboratory for Translational Genetics, Department of OncologyUniversity of LeuvenBelgium
| | - Ignace Vergote
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology and Leuven Cancer InstituteUniversity Hospitals LeuvenLeuvenBelgium
| | - Adriaan Vanderstichele
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology and Leuven Cancer InstituteUniversity Hospitals LeuvenLeuvenBelgium
| | - Els Van Nieuwenhuysen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology and Leuven Cancer InstituteUniversity Hospitals LeuvenLeuvenBelgium
| | - Hoda Anton‐Culver
- Department of Epidemiology, Director of Genetic Epidemiology Research Institute, Center for Cancer Genetics Research & Prevention, School of MedicineUniversity of California IrvineIrvineCalifornia
| | - Argyrios Ziogas
- Department of EpidemiologyUniversity of California IrvineIrvineCalifornia
| | - James D. Brenton
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Line Bjorge
- Department of Gynecology and ObstetricsHaukeland University HospitalBergenNorway
- Centre for Cancer Biomarkers, Department of Clinical ScienceUniversity of BergenBergenNorway
| | - Helga B. Salvensen
- Department of Gynecology and ObstetricsHaukeland University HospitalBergenNorway
- Centre for Cancer Biomarkers, Department of Clinical ScienceUniversity of BergenBergenNorway
| | - Lambertus A. Kiemeney
- Radboud University Medical CenterRadboud Institute for Health SciencesNijmegenNetherlands
| | - Leon F. A. G. Massuger
- Department of Gynaecology, Radboud University Medical CenterRadboud Institute for Molecular Life sciencesNijmegenThe Netherlands
| | - Tanja Pejovic
- Department of Obstetrics & GynecologyOregon Health & Science UniversityPortlandOregon
- Knight Cancer Institute, Oregon Health & Science UniversityPortlandOregon
| | - Amanda Bruegl
- Department of Obstetrics & GynecologyOregon Health & Science UniversityPortlandOregon
- Knight Cancer Institute, Oregon Health & Science UniversityPortlandOregon
| | - Melissa Moffitt
- Department of Obstetrics & GynecologyOregon Health & Science UniversityPortlandOregon
- Knight Cancer Institute, Oregon Health & Science UniversityPortlandOregon
| | - Linda Cook
- Division of Epidemiology and Biostatistics, Department of Internal MedicineUniversity of New MexicoAlbuquerqueNew Mexico
| | - Nhu D. Le
- Cancer Control Research, British Columbia Cancer AgencyVancouverBritish ColumbiaCanada
| | - Angela Brooks‐Wilson
- Canada's Michael Smith Genome Sciences CentreBritish Columbia Cancer AgencyVancouverBritish ColumbiaCanada
- Department of Biomedical Physiology and KinesiologySimon Fraser UniversityBurnabyBritish ColumbiaCanada
| | - Linda E. Kelemen
- Hollings Cancer Center and Department of Public Health SciencesMedical University of South CarolinaCharlestonSouth Carolina
| | - Paul D.P. Pharoah
- Strangeways Research laboratory, Department of Oncology, Department of Public Health and Primary CareUniversity of CambridgeCambridgeUK
| | - Honglin Song
- Strangeways Research Laboratory, Department of OncologyUniversity of CambridgeCambridgeUK
| | - Ian Campbell
- Cancer Genetics Laboratory, Research DivisionPeter MacCallum Cancer CentreVictoriaAustralia
- Department of PathologyUniversity of MelbourneParkvilleVictoriaAustralia
| | - Diana Eccles
- Faculty of MedicineUniversity of SouthamptonSouthamptonUK
| | - Anna DeFazio
- Centre for Cancer ResearchThe Westmead Institute for Medical Research, The University of SydneySydneyNew South WalesAustralia
- Department of Gynaecological OncologyWestmead HospitalSydneyNew South WalesAustralia
| | - Catherine J. Kennedy
- Centre for Cancer ResearchThe Westmead Institute for Medical Research, The University of SydneySydneyNew South WalesAustralia
- Department of Gynaecological OncologyWestmead HospitalSydneyNew South WalesAustralia
| | | |
Collapse
|
12
|
Olender J, Lee NH. Role of Alternative Splicing in Prostate Cancer Aggressiveness and Drug Resistance in African Americans. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1164:119-139. [PMID: 31576545 PMCID: PMC6777849 DOI: 10.1007/978-3-030-22254-3_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alternative splicing, the process of removing introns and joining exons of pre-mRNA, is critical for growth, development, tissue homeostasis, and species diversity. Dysregulation of alternative splicing can initiate and drive disease. Aberrant alternative splicing has been shown to promote the "hallmarks of cancer" in both hematological and solid cancers. Of interest, recent work has focused on the role of alternative splicing in prostate cancer and prostate cancer health disparities. We will provide a review of prostate cancer health disparities involving the African American population, alternative RNA splicing, and alternative splicing in prostate cancer. Lastly, we will summarize our work on differential alternative splicing in prostate cancer disparities and its implications for disparate health outcomes and therapeutic targets.
Collapse
Affiliation(s)
- Jacqueline Olender
- Department of Pharmacology and Physiology, GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Norman H Lee
- Department of Pharmacology and Physiology, GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
13
|
Burton LJ, Hawsawi O, Loyd Q, Henderson V, Howard S, Harlemon M, Ragin C, Roberts R, Bowen N, Gacii A, Odero-Marah V. Association of Epithelial Mesenchymal Transition with prostate and breast health disparities. PLoS One 2018; 13:e0203855. [PMID: 30199553 PMCID: PMC6130866 DOI: 10.1371/journal.pone.0203855] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022] Open
Abstract
African Americans (AA) have higher death rates due to prostate and breast cancer as compared to Caucasian Americans (CA), and few biomarkers have been associated with this disparity. In our study we investigated whether epithelial-mesenchymal transition (EMT) with a focus on Snail and Cathepsin L (Cat L), could potentially be two markers associated with prostate and breast health disparities. We have previously shown that Snail can increase Cat L protein and activity in prostate and breast cancer. Western blot and real-time PCR analyses showed that mesenchymal protein expression (Snail, vimentin, Cat L) and Cat L activity (shown by zymography) was higher in AA prostate cancer cells as compared to CA normal transformed RWPE-1 prostate epithelial cells, and androgen-dependent cells, and comparable to metastatic CA cell lines. With respect to breast cancer, mesenchymal markers were higher in TNBC compared to non-TNBC cells. The higher mesenchymal marker expression was functionally associated with higher proliferative and migratory rates. Immunohistochemistry showed that both nuclear Snail and Cat L expression was significantly higher in cancer compared to normal for CA and Bahamas prostate patient tissue. Interestingly, AA normal tissue stained higher for nuclear Snail and Cat L that was not significantly different to cancer tissue for both prostate and breast tissue, but was significantly higher than CA normal tissue. AA TNBC tissue also displayed significantly higher nuclear Snail expression compared to CA TNBC, while no significant differences were observed with Luminal A cancer tissue. Therefore, increased EMT in AA compared to CA that may contribute to the more aggressive disease.
Collapse
Affiliation(s)
- Liza J. Burton
- Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, United States of America
| | - Ohuod Hawsawi
- Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, United States of America
| | - Quentin Loyd
- Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, United States of America
| | - Veronica Henderson
- Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, United States of America
| | - Simone Howard
- Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, United States of America
| | - Maxine Harlemon
- Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, United States of America
- African Caribbean Cancer Consortium, Philadelphia, PA, United States of America
| | - Camille Ragin
- African Caribbean Cancer Consortium, Philadelphia, PA, United States of America
- Fox Chase Cancer Center, Temple Health, Philadelphia, PA, United States of America
| | - Robin Roberts
- African Caribbean Cancer Consortium, Philadelphia, PA, United States of America
- University of West Indies School of Clinical Medicine and Research, Nassau, The Bahamas
| | - Nathan Bowen
- Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, United States of America
| | - Andrew Gacii
- African Caribbean Cancer Consortium, Philadelphia, PA, United States of America
- Department of Lab Medicine, Kenyatta National Hospital, Nairobi, Kenya
| | - Valerie Odero-Marah
- Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, United States of America
- African Caribbean Cancer Consortium, Philadelphia, PA, United States of America
| |
Collapse
|
14
|
Wallace TJ, Qian J, Avital I, Bay C, Man YG, Wellman LL, Moskaluk C, Troyer D, Ramnani D, Stojadinovic A. Technical Feasibility of Tissue Microarray (TMA) Analysis of Tumor-Associated Immune Response in Prostate Cancer. J Cancer 2018; 9:2191-2202. [PMID: 29937939 PMCID: PMC6010688 DOI: 10.7150/jca.22846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 04/28/2018] [Indexed: 11/22/2022] Open
Abstract
Introduction: The androgen receptor (AR) regulates immune-related epithelial-to-mesenchymal transition (EMT), and prostate cancer (PCa) metastasis. Primary tumor-infiltrating lymphocytes (TILs) [CD3+, CD4+, and CD8+ TILs] are potential prognostic indicators in PCa, and variations may contribute to racial disparities in tumor biology and PCa outcomes. Aim: To assess the technical feasibility of tumor microarray (TMA)-based methods to perform multi-marker TIL profiling in primary resected PCa. Methods: Paraffin-embedded tissue cores of histopathologically-confirmed primary PCa (n = 40; 1 TMA tissue specimen loss) were arrayed in triplicate on TMAs. Expression profiles of AR, CD3+, CD4+, and CD8+ TILs in normal prostate, and the center and periphery of both the tumor-dominant nodule and highest Gleason grade were detected by IHC and associated with clinical and pathological data using standard statistical methodology. An independent pathologist, blinded to the clinical data, scored all samples (percent and intensity of positive cells). Results: TMAs were constructed from 21 (53.8%) Black and 18 (46.2%) White males with completely-resected, primarily pT2 stage PCa [pT2a (n = 3; 7.7%); pT2b (n = 2; 5.1%); pT2c (n = 27; 69.2%); pT3a (n = 5; 12.8%); mean pre-op PSA = 8.17 ng/ml]. The CD3, CD4, CD8, and CD8/CD3 cellular protein expression differed from normal in the periphery of the dominant nodule, the center of the highest Gleason grade, and the periphery of the highest Gleason grade (P < 0.05). Correlations between TIL expression in the center and periphery of the dominant nodule, with corresponding center and periphery of the highest Gleason grade, respectively, were robust, and the magnitude of these correlations differed markedly by race (P < 0.05). Conclusions: Multi-marker (AR, CD3, CD4, CD8) profiling with IHC analysis of TMAs consisting of primary, non-metastatic resected prostate cancer is technically feasible in this pilot study. Future studies will evaluate primary tumor immunoscore using semi-quantitative, IHC-based methodology to assess differences in the spectrum, quantity, and/or localization of TILs, and to gain insights into racial disparities in PCa tumor biology and clinical outcomes.
Collapse
Affiliation(s)
| | - Junqi Qian
- Virginia Urology, Richmond, Virginia, U.S.A
| | - Itzhak Avital
- Soroka University Center for Advanced Cancer Care, Ber Sheva, Israel
| | - Curt Bay
- A.T. Still University, Mesa, Arizona, U.S.A
| | - Yan-Gao Man
- National Medical Centre of Colorectal Disease, Third Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (TCM), Nanjing, China
| | | | - Chris Moskaluk
- University of Virginia, Charlottesville, Virginia, U.S.A
| | - Dean Troyer
- Eastern Virginia Medical School, Norfolk, Virginia, U.S.A
| | | | | |
Collapse
|
15
|
Kumar S, Singh R, Malik S, Manne U, Mishra M. Prostate cancer health disparities: An immuno-biological perspective. Cancer Lett 2018; 414:153-165. [PMID: 29154974 PMCID: PMC5743619 DOI: 10.1016/j.canlet.2017.11.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/09/2017] [Accepted: 11/11/2017] [Indexed: 02/07/2023]
Abstract
Prostate cancer (PCa) is the most commonly diagnosed malignancy in males, and, in the United States, is the second leading cause of cancer-related death for men older than 40 years. There is a higher incidence of PCa for African Americans (AAs) than for European-Americans (EAs). Investigations related to the incidence of PCa-related health disparities for AAs suggest that there are differences in the genetic makeup of these populations. Other differences are environmentally induced (e.g., diet and lifestyle), and the exposures are different. Men who immigrate from Eastern to Western countries have a higher risk of PCa than men in their native countries. However, the number of immigrants developing PCa is still lower than that of men in Western countries, suggesting that genetic factors are involved in the development of PCa. Altered genetic polymorphisms are associated with PCa progression. Androgens and the androgen receptor (AR) are involved in the development and progression of PCa. For populations with diverse racial/ethnic backgrounds, differences in lifestyle, diet, and biology, including genetic mutations/polymorphisms and levels of androgens and AR, are risk factors for PCa. Here, we provide an immuno-biological perspective on PCa in relation to racial/ethnic disparities and identify factors associated with the disproportionate incidence of PCa and its clinical outcomes.
Collapse
Affiliation(s)
- Sanjay Kumar
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, Montgomery, AL 36104, USA
| | - Rajesh Singh
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Shalie Malik
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, Montgomery, AL 36104, USA; Department of Zoology, University of Lucknow, Lucknow 226007, India
| | - Upender Manne
- Department of Pathology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Manoj Mishra
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, Montgomery, AL 36104, USA.
| |
Collapse
|
16
|
Smith CJ, Minas TZ, Ambs S. Analysis of Tumor Biology to Advance Cancer Health Disparity Research. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:304-316. [PMID: 29137948 DOI: 10.1016/j.ajpath.2017.06.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/24/2017] [Accepted: 06/29/2017] [Indexed: 12/20/2022]
Abstract
Cancer mortality rates in the United States continue to decline. Reductions in tobacco use, uptake of preventive measures, adoption of early detection methods, and better treatments have resulted in improved cancer outcomes for men and women. Despite this progress, some population groups continue to experience an excessive cancer burden when compared with other population groups. One of the most prominent cancer health disparities exists in prostate cancer. Prostate cancer mortality rates are highest among men of African ancestry when compared with other men, both in the United States and globally. This disparity and other cancer health disparities are largely explained by differences in access to health care, diet, lifestyle, cultural barriers, and disparate exposures to carcinogens and pathogens. Dietary and lifestyle factors, pathogens, and ancestry-related factors can modify tumor biology and induce a more aggressive disease. There are numerous examples of how environmental exposures, like tobacco, chronic stress, or dietary factors, induce an adverse tumor biology, leading to a more aggressive disease and decreased patient survival. Because of population differences in the exposure to these risk factors, they can be the cause of cancer disparities. In this review, we will summarize recent advances in our understanding of prostate and breast cancer disparities in the United States and discuss how the analysis of tumor biology can advance health disparity research.
Collapse
Affiliation(s)
- Cheryl J Smith
- Laboratory of Human Carcinogenesis, Center of Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Tsion Z Minas
- Laboratory of Human Carcinogenesis, Center of Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center of Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
17
|
Sathekge M, Lengana T, Maes A, Vorster M, Zeevaart J, Lawal I, Ebenhan T, Van de Wiele C. 68Ga-PSMA-11 PET/CT in primary staging of prostate carcinoma: preliminary results on differences between black and white South-Africans. Eur J Nucl Med Mol Imaging 2017; 45:226-234. [PMID: 29101444 PMCID: PMC5745572 DOI: 10.1007/s00259-017-3852-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/26/2017] [Indexed: 12/15/2022]
Abstract
PURPOSE The incidence of prostate cancer is 60% higher and the mortality rate is two- to three-times greater in black versus white men. We report on differences in 68Ga-PSMA-11 PET/CT imaging findings in 77 black South-African (BSAs) and 18 white South-African (WSAs) treatment-naïve primary prostate carcinoma (PPC) patients. METHODS 68Ga-PSMA-11 PET/CT imaging findings were compared to histological, biochemical and morphological imaging data. Patients were grouped into three Gleason grade groups (GG), GG 1 (scores 3 + 3 and 3 + 4), GG2 (scores 4 + 3 and 4 + 4) and GG3 (scores 9 and 10), and the PSA difference among the groups was determined. Inter-racial difference in SUVmax of the primary tumor as well as its correlation with serum PSA were also determined. RESULTS Ninety-three out of 95 PPC where readily identified on 68Ga-PSMA-11 PET/CT imaging. Median PPC SUVmax and serum PSA values proved significantly higher (p = 0.033 and p = 0.003) in GG3 patients (median 16.4 and 180 ng/ml) when compared to GG1 patients (median 9.6 and 25.1 ng/ml) or GG2 patients (median 8.8 and 46.2 ng/ml). SUVmax significantly correlated with serum PSA-values (r = 0.377 (p = 0.0001)). Age, frequency of lymph node involvement and distant metastases, and GGs (p ≥ 0.153) were similar in BSAs and WSAs, both median serum PSA-values as well as SUVmax values proved significantly higher in BSAs when compared to WSAs, respectively, 81.6 ng/ml versus 14.5 ng/ml (p = 0.0001) and 11.9 versus 4.38 (p = 0.004). Moreover, Gleason-score normalized median SUVmax values proved 2.5 times higher in BSAs when compared to WSAs (p = 0.005). CONCLUSION SUVmax values proved significantly related to GG and to be significantly higher in BSAs when compared to WSAs. Also, SUVmax significantly correlated with serum PSA values, which was significantly higher in BSAs when compared with WSAs.
Collapse
Affiliation(s)
- Mike Sathekge
- Department of Nuclear medicine, University of Pretoria, Pretoria, South Africa. .,Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001, South Africa.
| | - Thabo Lengana
- Department of Nuclear medicine, University of Pretoria, Pretoria, South Africa
| | - Alex Maes
- Department of Nuclear medicine, University of Pretoria, Pretoria, South Africa.,Department of Nuclear Medicine, AZ Groeninge, Kortrijk, Belgium.,Department of Nuclear Medicine and Pathology, KULAK, Kortrijk, Belgium
| | - Mariza Vorster
- Department of Nuclear medicine, University of Pretoria, Pretoria, South Africa
| | - JanRijn Zeevaart
- Department of Nuclear medicine, University of Pretoria, Pretoria, South Africa.,Radiochemistry, The South African Nuclear Energy Corporation SOC Ltd (Necsa), Pelindaba, Pretoria, South Africa
| | - Ismaheel Lawal
- Department of Nuclear medicine, University of Pretoria, Pretoria, South Africa
| | - Thomas Ebenhan
- Department of Nuclear medicine, University of Pretoria, Pretoria, South Africa
| | - Christophe Van de Wiele
- Department of Nuclear medicine, University of Pretoria, Pretoria, South Africa.,Department of Nuclear Medicine, AZ Groeninge, Kortrijk, Belgium.,Department of Radiology and Nuclear Medicine, University Ghent, Ghent, Belgium
| |
Collapse
|
18
|
Wang Y, Freedman JA, Liu H, Moorman PG, Hyslop T, George DJ, Lee NH, Patierno SR, Wei Q. Associations between RNA splicing regulatory variants of stemness-related genes and racial disparities in susceptibility to prostate cancer. Int J Cancer 2017; 141:731-743. [PMID: 28510291 DOI: 10.1002/ijc.30787] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/24/2017] [Accepted: 05/02/2017] [Indexed: 01/01/2023]
Abstract
Evidence suggests that cells with a stemness phenotype play a pivotal role in oncogenesis, and prostate cells exhibiting this phenotype have been identified. We used two genome-wide association study (GWAS) datasets of African descendants, from the Multiethnic/Minority Cohort Study of Diet and Cancer (MEC) and the Ghana Prostate Study, and two GWAS datasets of non-Hispanic whites, from the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial and the Breast and Prostate Cancer Cohort Consortium (BPC3), to analyze the associations between genetic variants of stemness-related genes and racial disparities in susceptibility to prostate cancer. We evaluated associations of single-nucleotide polymorphisms (SNPs) in 25 stemness-related genes with prostate cancer risk in 1,609 cases and 2,550 controls of non-Hispanic whites (4,934 SNPs) and 1,144 cases and 1,116 controls of African descendants (5,448 SNPs) with correction by false discovery rate ≤0.2. We identified 32 SNPs in five genes (TP63, ALDH1A1, WNT1, MET and EGFR) that were significantly associated with prostate cancer risk, of which six SNPs in three genes (TP63, ALDH1A1 and WNT1) and eight EGFR SNPs showed heterogeneity in susceptibility between these two racial groups. In addition, 13 SNPs in MET and one in ALDH1A1 were found only in African descendants. The in silico bioinformatics analyses revealed that EGFR rs2072454 and SNPs in linkage with the identified SNPs in MET and ALDH1A1 (r2 > 0.6) were predicted to regulate RNA splicing. These variants may serve as novel biomarkers for racial disparities in prostate cancer risk.
Collapse
Affiliation(s)
- Yanru Wang
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC
| | - Jennifer A Freedman
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC
| | - Patricia G Moorman
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Community and Family Medicine, Duke University Medical Center, Durham, NC
| | - Terry Hyslop
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC
| | - Daniel J George
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC
| | - Norman H Lee
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Steven R Patierno
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC.,Department of Community and Family Medicine, Duke University Medical Center, Durham, NC.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC
| |
Collapse
|
19
|
Kumar NB, Pow-Sang JM, Spiess PE, Park JY, Chornokur G, Leone AR, Phelan CM. Chemoprevention in African American Men With Prostate Cancer. Cancer Control 2017; 23:415-423. [PMID: 27842331 DOI: 10.1177/107327481602300413] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Recommendations for cancer screening are uncertain for the early detection or prevention of prostate cancer in African American men. Thus, chemoprevention strategies are needed to specifically target African American men. METHODS The evidence was examined on the biological etiology of disparities in African Americans related to prostate cancer. Possible chemopreventive agents and biomarkers critical to prostate cancer in African American men were also studied. RESULTS High-grade prostatic intraepithelial neoplasia may be more prevalent in African American men, even after controlling for age, prostate-specific antigen (PSA) level, abnormal results on digital rectal examination, and prostate volume. Prostate cancer in African American men can lead to the overexpression of signaling receptors that may mediate increased proliferation, angiogenesis, and decreased apoptosis. Use of chemopreventive agents may be useful for select populations of men. CONCLUSIONS Green tea catechins are able to target multiple pathways to address the underlying biology of prostate carcinogenesis in African American men, so they may be ideal as a chemoprevention agent in these men diagnosed with high-grade prostatic intraepithelial neoplasia.
Collapse
Affiliation(s)
- Nagi B Kumar
- Department of Epidemiology, Moffitt Cancer Center, Tampa, FL, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
Bhardwaj A, Srivastava SK, Khan MA, Prajapati VK, Singh S, Carter JE, Singh AP. Racial disparities in prostate cancer: a molecular perspective. Front Biosci (Landmark Ed) 2017; 22:772-782. [PMID: 27814645 DOI: 10.2741/4515] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Prostate cancer incidence and mortality rates are remarkably higher in African-American men as compared to their European-Americans counterparts. Despite these recognitions, precise causes underlying such prevalent racial disparities remain poorly understood. Although socioeconomic factors could account for such differences up to a certain extent, it is now being increasingly realized that such disparity has a molecular basis. Indeed, several differences, including genetic polymorphism, gene mutations, epigenetic modifications, miRNAs alterations, etc., have been reported in malignant prostate tissues from patients of diverse racial backgrounds. Here, we attempt to provide a molecular perspective on prostate cancer racial disparities by gathering available information on these associated factors and discussing their potential significance in disproportionate incidence and clinical outcomes.
Collapse
Affiliation(s)
- Arun Bhardwaj
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, 1660 Spring Hill Avenue, Mobile-36604-1405, Alabama, USA,
| | - Sanjeev K Srivastava
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Mohammad Aslam Khan
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Vijay K Prajapati
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Seema Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - James E Carter
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Ajay P Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
21
|
Singh SK, Lillard JW, Singh R. Molecular basis for prostate cancer racial disparities. Front Biosci (Landmark Ed) 2017; 22:428-450. [PMID: 27814623 DOI: 10.2741/4493] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Prostate cancer (PCa) remains the most common cancer in American men. African-American (AA) men continue to have higher PCa prevalence and mortality rates compared to men in other populations. In addition to socioeconomic factors and lifestyle differences, molecular alterations contribute to this discrepancy. We summarize molecular genetics research results interrelated with the biology of PCa racial disparity. Androgen and androgen receptor (AR) pathways have long been associated with prostate growth. Racial differences have also been found among variants of genes of the enzymes involved in androgen biosynthesis and metabolism. Growth factors and their receptors are a potential cause of the disparity in PCa. Recent molecular and biotechnological approaches in the field of proteomics and genomics will greatly aid the advancement of translational research on racial disparity in PCa, which may help, in finding new prognostic markers and novel therapeutic approaches for the treatment of PCa in AA.
Collapse
Affiliation(s)
- Santosh K Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - James W Lillard
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Rajesh Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia 30310,
| |
Collapse
|
22
|
Zhang L, Wang J, Wang Y, Zhang Y, Castro P, Shao L, Sreekumar A, Putluri N, Guha N, Deepak S, Padmanaban A, Creighton CJ, Ittmann M. MNX1 Is Oncogenically Upregulated in African-American Prostate Cancer. Cancer Res 2016; 76:6290-6298. [PMID: 27578002 DOI: 10.1158/0008-5472.can-16-0087] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 07/28/2016] [Indexed: 11/16/2022]
Abstract
Incidence and mortality rates for prostate cancer are higher in African-American (AA) men than in European-American (EA) men, but the biologic basis for this disparity is unclear. We carried out a detailed analysis of gene expression changes in prostate cancer compared with their matched benign tissues in a cohort of AA men and compared them with existing data from EA men. In this manner, we identified MNX1 as a novel oncogene upregulated to a relatively greater degree in prostate cancer from AA men. Androgen and AKT signaling play a central role in the pathogenesis of prostate cancer and we found that both of these signaling pathways increased MNX1 expression. MNX1 in turn upregulated lipid synthesis by stimulating expression of SREBP1 and fatty acid synthetase. Our results define MNX1 as a novel targetable oncogene increased in AA prostate cancer that is associated with aggressive disease. Cancer Res; 76(21); 6290-8. ©2016 AACR.
Collapse
Affiliation(s)
- Li Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China.,Department of Pathology and Immunology and Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - Jianghua Wang
- Department of Pathology and Immunology and Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - Yongquan Wang
- Department of Pathology and Immunology and Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - Yiqun Zhang
- Division of Biostatistics, Dan L. Duncan Cancer Center, Houston, Texas
| | - Patricia Castro
- Department of Pathology and Immunology and Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - Longjiang Shao
- Department of Pathology and Immunology and Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - Arun Sreekumar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.,Verna and Marrs McLean Department of Biochemistry, Baylor College of Medicine, Houston, Texas.,Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, Texas
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.,Verna and Marrs McLean Department of Biochemistry, Baylor College of Medicine, Houston, Texas.,Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, Texas
| | - Nilanjan Guha
- Agilent Technologies India Pvt. Ltd, Bangalore, India
| | | | | | - Chad J Creighton
- Division of Biostatistics, Dan L. Duncan Cancer Center, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Michael Ittmann
- Department of Pathology and Immunology and Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas.
| |
Collapse
|
23
|
Nelson CJ, Starr TD, Macchia RJ, Hyacinthe L, Friedman S, Roth AJ. Assessing anxiety in Black men with prostate cancer: further data on the reliability and validity of the Memorial Anxiety Scale for Prostate Cancer (MAX-PC). Support Care Cancer 2016; 24:2905-11. [PMID: 26847348 DOI: 10.1007/s00520-016-3092-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/17/2016] [Indexed: 11/29/2022]
Abstract
PURPOSE The National Cancer Institute has highlighted the need for psychosocial research to focus on Black cancer patients. This applies to Black men with prostate cancer, as there is little systematic research concerning psychological distress in these men. This study was designed to validate the Memorial Anxiety Scale for Prostate Cancer (MAX-PC) in Black men with prostate cancer to help facilitate research within this group. METHODS At three institutions, Black men with prostate cancer (n = 101) completed the MAX-PC, the Hospital Anxiety and Depression Scale (HADS), the Functional Assessment of Cancer Therapy (FACT) Quality of Life Questionnaire, and the Distress Thermometer. RESULTS The average age of the 101 men was 66 (SD = 10) and 58 % had early-stage disease. The MAX-PC and its subscales (Prostate Cancer Anxiety, PSA Anxiety, and Fear of Recurrence) produced strong coefficient alphas (0.89, 0.88, 0.71, and 0.77, respectively). Factor analysis supported the three-factor structure of the scale established in earlier findings. The MAX-PC also demonstrated strong validity. MAX-PC total scores correlated highly with the Anxiety subscale of the HADS (r = 0.59, p < 0.01) and the FACT Emotional Well-Being subscale (r = -0.55, p < 0.01). Demonstrating discriminant validity, the correlation with the HADS Depression subscale (r = 0.40, p < 0.01) and the CES-D (r = 0.42, p < 0.01) was lower compared to that with the HADS Anxiety subscale. CONCLUSIONS The MAX-PC is valid and reliable in Black men with prostate cancer. We hope the validation of this scale in Black men will help facilitate psychosocial research in this group that is disproportionately adversely affected by this cancer.
Collapse
Affiliation(s)
- Christian J Nelson
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, 641 Lexington Ave., 7th Floor, New York, NY, 10022, USA.
| | - Tatiana D Starr
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, 641 Lexington Ave., 7th Floor, New York, NY, 10022, USA
| | - Richard J Macchia
- Department of Urology, Cleveland Clinic, Weston, FL, USA.,Department of Urology, Kings County Hospital Center, Brooklyn, NY, USA.,Department of Urology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | | | - Steven Friedman
- Department of Psychiatry, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Andrew J Roth
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, 641 Lexington Ave., 7th Floor, New York, NY, 10022, USA
| |
Collapse
|
24
|
Wang BD, Ceniccola K, Yang Q, Andrawis R, Patel V, Ji Y, Rhim J, Olender J, Popratiloff A, Latham P, Lai Y, Patierno SR, Lee NH. Identification and Functional Validation of Reciprocal microRNA-mRNA Pairings in African American Prostate Cancer Disparities. Clin Cancer Res 2015; 21:4970-84. [PMID: 26089375 DOI: 10.1158/1078-0432.ccr-14-1566] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 06/08/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE African Americans (AA) exhibit higher rates of prostate cancer incidence and mortality compared with European American (EA) men. In addition to socioeconomic influences, biologic factors are believed to play a critical role in prostate cancer disparities. We investigated whether population-specific and -enriched miRNA-mRNA interactions might contribute to prostate cancer disparities. EXPERIMENTAL DESIGN Integrative genomics was used, combining miRNA and mRNA profiling, miRNA target prediction, pathway analysis, and functional validation, to map miRNA-mRNA interactions associated with prostate cancer disparities. RESULTS We identified 22 AA-specific and 18 EA-specific miRNAs in prostate cancer versus patient-matched normal prostate, and 10 "AA-enriched/-depleted" miRNAs in AA prostate cancer versus EA prostate cancer comparisons. Many of these population-specific/-enriched miRNAs could be paired with target mRNAs that exhibited an inverse pattern of differential expression. Pathway analysis revealed EGFR (or ERBB) signaling as a critical pathway significantly regulated by AA-specific/-enriched mRNAs and miRNA-mRNA pairings. Novel miRNA-mRNA pairings were validated by qRT-PCR, Western blot, and/or IHC analyses in prostate cancer specimens. Loss/gain of function assays performed in population-specific prostate cancer cell lines confirmed miR-133a/MCL1, miR-513c/STAT1, miR-96/FOXO3A, miR-145/ITPR2, and miR-34a/PPP2R2A as critical miRNA-mRNA pairings driving oncogenesis. Manipulating the balance of these pairings resulted in decreased proliferation and invasion, and enhanced sensitization to docetaxel-induced cytotoxicity in AA prostate cancer cells. CONCLUSIONS Our data suggest that AA-specific/-enriched miRNA-mRNA pairings may play a critical role in the activation of oncogenic pathways in AA prostate cancer. Our findings also suggest that miR-133a/MCL1, miR-513c/STAT1, and miR-96/FOXO3A may have clinical significance in the development of novel strategies for treating aggressive prostate cancer.
Collapse
Affiliation(s)
- Bi-Dar Wang
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Kristin Ceniccola
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Qi Yang
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Ramez Andrawis
- Medical Faculty Associates, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Vyomesh Patel
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
| | - Youngmi Ji
- Cartilage Biology and Orthopedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | - Johng Rhim
- Department of Surgery, Center for Prostate Disease Research, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Jacqueline Olender
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Anastas Popratiloff
- Department of Anatomy and Regenerative Biology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Patricia Latham
- Department of Pathology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Yinglei Lai
- Department of Statistics, The George Washington University, Washington, District of Columbia
| | - Steven R Patierno
- GW Cancer Institute, The George Washington University Medical Center, Washington, District of Columbia. Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| | - Norman H Lee
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia.
| |
Collapse
|
25
|
A Population-Based Study of Men With Low-Volume Low-Risk Prostate Cancer: Does African-American Race Predict for More Aggressive Disease? Clin Genitourin Cancer 2015; 13:e259-e264. [PMID: 25777681 DOI: 10.1016/j.clgc.2015.02.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/29/2015] [Accepted: 02/16/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND Because of recent reports that suggested more pathologically aggressive disease in African-American (AA) men, we sought to compare pathologic features between AA and Caucasian-American men with low-risk, low-volume prostate cancer. MATERIALS AND METHODS We analyzed the Surveillance, Epidemiology, and End Results database for pathologic differences based on race. Data on all men who were diagnosed between 2010 and 2011 with prostate cancer, T1cN0M0, Gleason score of 6 (3+3), prostate-specific antigen < 10 ng/mL, via a 12-core biopsy and had ≤ 2 positive samples, and underwent radical prostatectomy were abstracted. Univariate and multivariate logistic regression were performed to detect predictors for adverse pathology, which was primarily defined as pT2 and Gleason ≥ 4+3, or pT3a and Gleason 3+3 with positive margins, pT3a and Gleason ≥ 3+4, or pT3b-pT4 with any Gleason score. RESULTS There were 1794 men who met the target study criteria. AA men were a median of 3 years younger (P < .001), and were more likely to have 2 positive cores (P = .02). However, there were no statistically significant differences between Caucasian and AA men regarding pathologic Gleason score (P = .99), pathologic extent of disease (P = .34), margins (P = .43), Cancer of the Prostate Risk Assessment score (P = .56), or adverse features (P = .45). On multivariate analysis, there were no differences between AA and Caucasian men with regard to adverse pathologic features (odds ratio, 1.43; 95% confidence interval, 0.87-1.24; P = .16). CONCLUSION In the absence of definitive data to support a more aggressive natural history of very low risk prostate cancer in AA men, these data support continued use of active surveillance in this population.
Collapse
|
26
|
Reams RR, Jones-Triche J, Chan OTM, Hernandez BY, Soliman KFA, Yates C. Immunohistological analysis of ABCD3 expression in Caucasian and African American prostate tumors. BIOMED RESEARCH INTERNATIONAL 2015; 2015:132981. [PMID: 25802834 PMCID: PMC4329846 DOI: 10.1155/2015/132981] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 01/04/2015] [Indexed: 01/13/2023]
Abstract
In a previously published study, we showed that expression of the ABCD3 gene increased with increasing metastatic potential in a panel of prostate cancer cell lines derived from African American and Caucasian American men. Given importance of identifying biomarker(s) that can distinguish indolent versus aggressive prostate tumors, we conducted an immunohistochemical analysis of ABCD3 expression Caucasian and African American prostate tumors. ABCD3 expression in each patient population was compared with clinicopathologic characteristics, Gleason score, and age. ABCD3 expression increased with increasing Gleason score (P = 0.0094), age (P = 0.0014), and pathology grade (P = 0.0007) in Caucasian patients. Interestingly, in the AA patients, ABCD3 expression highly increased to the same degree in both low and high Gleason score tumors. Similarly, ABCD3 expression was elevated to the same degree in BPH derived from AA. Our findings demonstrate that increased ABCD3 expression correlates with Gleason Score in CA prostate tumors. However, in AA prostate tumors, ABCD3 expression was higher and was sustained in both low Gleason and high Gleason AA tumors. While the functional role of ABCD3 in prostate cancer is not completely elucidated, this gene warrants further study as a potential biomarker for aggressive prostate.
Collapse
Affiliation(s)
- R. Renee Reams
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | | | - Owen T. M. Chan
- Cancer Epidemiology, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Brenda Y. Hernandez
- Cancer Epidemiology, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Karam F. A. Soliman
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Clayton Yates
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, AL 36088, USA
| |
Collapse
|
27
|
Poku RA, Amissah F, Duverna R, Aguilar BJ, Kiros GE, Lamango NS. Polyisoprenylated methylated protein methyl esterase as a putative drug target for androgen-insensitive prostate cancer. Ecancermedicalscience 2014; 8:459. [PMID: 25228915 PMCID: PMC4154947 DOI: 10.3332/ecancer.2014.459] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Indexed: 02/05/2023] Open
Abstract
Prostate cancer (CaP) is the most frequently diagnosed cancer in US men, with an estimated 236,590 new cases and 29,720 deaths in 2013. There exists the need to identify biomarkers/therapeutic targets for the early/companion diagnosis and development of novel therapies against the recalcitrant disease. Mutation and overexpression-induced abnormal activities of polyisoprenylated proteins have been implicated in CaP. Polyisoprenylated methylated protein methyl esterase (PMPMEase) catalyses the only reversible and terminal reaction of the polyisoprenylation pathway and may promote the effects of G proteins on cell viability. In this review, the potential role of PMPMEase to serve as a new drug target for androgen-insensitive CaP was determined. Specific PMPMEase activities were found to be 3.5- and 4.5-fold higher in androgen-sensitive 22Rv1 and androgen-dependent LNCaP and 1.5- and 9.8-fold higher in castration-resistant DU 145 and PC-3 CaP cells compared to normal WPE1-NA22 prostate cells. The PMPMEase inhibitor, L-28, induced apoptosis with EC50 values ranging from 1.8 to 4.6 μM. The PMPMEase activity in the cells following treatment with L-28 followed a similar profile, with IC50 ranging from 2.3 to 130 μM. L-28 disrupted F-actin filament organisation at 5 μM and inhibited cell migration 4-fold at 2 μM. Analysis of a CaP tissue microarray for PMPMEase expression revealed intermediate, strong, and very strong staining in 94.5% of the 92 adenocarcinoma cases compared to trace and weak staining in the normal and normal-adjacent tissue controls. The data are an indication that effective targeting of PMPMEase through the development of more potent agents may lead to the successful treatment of metastatic CaP.
Collapse
Affiliation(s)
- Rosemary A Poku
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| | - Felix Amissah
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| | - Randolph Duverna
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| | - Byron J Aguilar
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| | - Gebre-Egziabher Kiros
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| | - Nazarius S Lamango
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| |
Collapse
|
28
|
WANG LIJUAN, HAN SUXIA, JIN GUIHUA, ZHOU XIA, LI MENG, YING XIA, WANG LE, WU HUILI, ZHU QING. Linc00963: A novel, long non-coding RNA involved in the transition of prostate cancer from androgen-dependence to androgen-independence. Int J Oncol 2014; 44:2041-9. [DOI: 10.3892/ijo.2014.2363] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 03/04/2014] [Indexed: 11/06/2022] Open
|
29
|
Jones J, Grizzle W, Wang H, Yates C. MicroRNAs that affect prostate cancer: emphasis on prostate cancer in African Americans. Biotech Histochem 2013; 88:410-24. [PMID: 23901944 DOI: 10.3109/10520295.2013.807069] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Although concerted efforts have been directed toward eradicating health disparities in the United States, the disease and mortality rates for African American men still are among the highest in the world. We focus here on the role of microRNAs (miRNAs) in the signaling pathways of androgen receptors and growth factors that promote the progression of prostate cancer to more aggressive disease. We explore also how differential expression of miRNAs contributes to aggressive prostate cancer including that of African Americans.
Collapse
Affiliation(s)
- J Jones
- Department of Biology and Center for Cancer Research, Tuskegee University , Tuskegee, Alabama
| | | | | | | |
Collapse
|
30
|
Prostate cancer chemoprevention in men of African descent: current state of the art and opportunities for future research. Cancer Causes Control 2013; 24:1465-80. [PMID: 23737026 DOI: 10.1007/s10552-013-0241-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 05/25/2013] [Indexed: 01/13/2023]
Abstract
Prostate cancer is the most frequently diagnosed malignancy in men. However, African American/Black men are 60 % more likely to be diagnosed with and 2.4 times more likely to die from prostate cancer, compared to Non-Hispanic White men. Despite the increased burden of this malignancy, no evidence-based recommendation regarding prostate cancer screening exists for the high-risk population. Moreover, in addition to screening and detection, African American men may constitute a prime population for chemoprevention. Early detection and chemoprevention may thus represent an integral part of prostate cancer control in this population. Importantly, recent research has elucidated biological differences in the prostate tumors of African American compared to European American men. The latter may enable a more favorable response in African American men to specific chemopreventive agents that target relevant signal transduction pathways. Based on this evolving evidence, the aims of this review are threefold. First, we aim to summarize the biological differences that were reported in the prostate tumors of African American and European American men. Second, we will review the single- and multi-target chemopreventive agents placing specific emphasis on the pathways implicated in prostate carcinogenesis. And lastly, we will discuss the most promising nutraceutical chemopreventive compounds. Our review underscores the promise of chemoprevention in prostate cancer control, as well as provides justification for further investment in this filed to ultimately reduce prostate cancer morbidity and mortality in this high-risk population of African American men.
Collapse
|
31
|
Powell IJ, Bollig-Fischer A. Minireview: the molecular and genomic basis for prostate cancer health disparities. Mol Endocrinol 2013; 27:879-91. [PMID: 23608645 DOI: 10.1210/me.2013-1039] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Despite more aggressive screening across all demographics and gradual declines in mortality related to prostate cancer (PCa) in the United States, race disparities persist. For African American men (AAM), PCa is more often an aggressive disease showing increased metastases and greater PCa-related mortality compared with European American men. The earliest research points to how distinctions are likely the result of a combination of factors, including ancestry genetics and lifestyle variables. More recent research considers that cancer, although influenced by external forces, is ultimately a disease primarily driven by aberrations observed in the molecular genetics of the tumor. Research studying PCa predominantly from European American men shows that indolent and advanced or metastatic prostate tumors have distinguishing molecular genomic make-ups. Early yet increasing evidence suggests that clinically distinct PCa from AAM also display molecular distinctions. It is reasonable to predict that further study will reveal molecular subtypes and various frequencies for PCa subtypes among diverse patient groups, thereby providing insight as to the genomic lesions and gene signatures that are functionally implicated in carcinogenesis or aggressive PCa in AAM. That knowledge will prove useful in developing strategies to predict who will develop advanced PCa among AAM and will provide the rationale to develop effective individualized treatment strategies to overcome disparities.
Collapse
Affiliation(s)
- Isaac J Powell
- Barbara Ann Karmanos Cancer Institute, Detroit, Michigan 48201, USA.
| | | |
Collapse
|
32
|
Nuclear Kaiso indicates aggressive prostate cancers and promotes migration and invasiveness of prostate cancer cells. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1836-46. [PMID: 22974583 DOI: 10.1016/j.ajpath.2012.08.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 07/25/2012] [Accepted: 08/01/2012] [Indexed: 12/13/2022]
Abstract
Kaiso, a p120 catenin-binding protein, is expressed in the cytoplasmic and nuclear compartments of cells; however, the biological consequences and clinical implications of a shift between these compartments have yet to be established. Herein, we report an enrichment of nuclear Kaiso expression in cells of primary and metastatic prostate tumors relative to the normal prostate epithelium. Nuclear expression of Kaiso correlates with Gleason score (P < 0.001) and tumor grade (P < 0.001). There is higher nuclear expression of Kaiso in primary tumor/normal matched samples and in primary tumors from African American men (P < 0.0001). We further found that epidermal growth factor (EGF) receptor up-regulates Kaiso at the RNA and protein levels in prostate cancer cell lines, but more interestingly causes a shift of cytoplasmic Kaiso to the nucleus that is reversed by the EGF receptor-specific kinase inhibitor, PD153035. In both DU-145 and PC-3 prostate cancer cell lines, Kaiso inhibition (short hairpin RNA-Kaiso) decreased cell migration and invasion even in the presence of EGF. Further, Kaiso directly binds to the E-cadherin promoter, and inhibition of Kaiso in PC-3 cells results in increased E-cadherin expression, as well as re-establishment of cell-cell contacts. In addition, Kaiso-depleted cells show more epithelial morphology and a reversal of the mesenchymal markers N-cadherin and fibronectin. Our findings establish a defined oncogenic role of Kaiso in promoting the progression of prostate cancer.
Collapse
|
33
|
Abstract
It is becoming clear that some of the differences in cancer risk, incidence and survival among people of different racial and ethnic backgrounds can be attributed to biological factors. However, identifying these factors and exploiting them to help eliminate cancer disparities has proved challenging. With this in mind, we asked four scientists for their opinions on the most crucial advances, as well as the challenges and what the future holds for this important emerging area of research.
Collapse
Affiliation(s)
- Brian E. Henderson
- The Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, California 90033-0804, USA
| | - Norman H. Lee
- The George Washington University Medical Center, Washington, DC, Department of Pharmacology & Physiology, 2300 I Street Northwest, Ross Hall, Washington, DC 20037, USA
| | - Victoria Seewaldt
- Department of Medicine, Duke University, BOX 2628, Room 221A MSRB, DUMC, Durham, North Carolina 27710, USA
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics and Ministry of Education (MOE) Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, 140 Hanzhong Rd., Nanjing 210029, Jiangsu, China; is also at The Section of Clinical Epidemiology, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| |
Collapse
|
34
|
Koscuiszka M, Hatcher D, Christos PJ, Rose AE, Greenwald HS, Chiu YL, Taneja SS, Mazumdar M, Lee P, Osman I. Impact of race on survival in patients with clinically nonmetastatic prostate cancer who deferred primary treatment. Cancer 2012; 118:3145-52. [PMID: 22020835 PMCID: PMC3623265 DOI: 10.1002/cncr.26619] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 08/26/2011] [Accepted: 09/19/2011] [Indexed: 11/07/2022]
Abstract
BACKGROUND Prostate cancer (PCa) racial disparity studies typically focus on survival differences after curative treatment. The authors of this report hypothesized that comparing mortality rates between African American (AA) and Caucasian American (CA) patients who deferred primary treatment for clinically nonmetastatic PCa may provide a better assessment of the impact of race on the natural course of PCa. METHODS The pathology database of the New York Veterans Administration Medical Center (VAMC), an equal access-of-care facility, was searched for patients with biopsy-proven PCa. Inclusion criteria included 1) no evidence of metastatic disease or death within 3 years after diagnosis, 2) no primary treatment, and 3) a minimum of 5 years of follow-up for survivors. RESULTS In total, 518 patients met inclusion criteria between 1990 and 2005. AA patients were younger (P = .02) and had higher median prostate-specific antigen (PSA) levels (P = .001) at the time of diagnosis compared with CA patients. In a multivariate model, higher Gleason score and PSA level were associated with increased mortality (P = .001 and P = .03, respectively), but race was not a predictor of death from PCa. CONCLUSIONS The current data suggested that race did not have a major impact on survival in patients with PCa who deferred primary treatment for clinically nonmetastatic disease.
Collapse
Affiliation(s)
- Michael Koscuiszka
- Department of Urology, New York University School of Medicine, New York, New York
| | - David Hatcher
- Department of Urology, New York University School of Medicine, New York, New York
| | - Paul J. Christos
- Division of Biostatistics and Epidemiology, Weill Medical College of Cornell University, New York, New York
| | - Amy E. Rose
- Department of Urology, New York University School of Medicine, New York, New York
| | | | - Ya-lin Chiu
- Division of Biostatistics and Epidemiology, Weill Medical College of Cornell University, New York, New York
| | - Samir S. Taneja
- Department of Urology, New York University School of Medicine, New York, New York
| | - Madhu Mazumdar
- Division of Biostatistics and Epidemiology, Weill Medical College of Cornell University, New York, New York
| | - Peng Lee
- Department of Urology, New York University School of Medicine, New York, New York
- Department of Pathology, New York University School of Medicine, New York, New York
- New York Harbor Healthcare System, New York, New York
| | - Iman Osman
- Department of Urology, New York University School of Medicine, New York, New York
- New York University Cancer Institute, New York, New York
- New York Harbor Healthcare System, New York, New York
| |
Collapse
|
35
|
Fu W, Madan E, Yee M, Zhang H. Progress of molecular targeted therapies for prostate cancers. Biochim Biophys Acta Rev Cancer 2011; 1825:140-52. [PMID: 22146293 DOI: 10.1016/j.bbcan.2011.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 11/18/2011] [Accepted: 11/19/2011] [Indexed: 01/16/2023]
Abstract
Prostate cancer remains the most commonly diagnosed malignancy and the second leading cause of cancer-related deaths in men in the United States. The current standard of care consists of prostatectomy and radiation therapy, which may often be supplemented with hormonal therapies. Recurrence is common, and many develop metastatic prostate cancer for which chemotherapy is only moderately effective. It is clear that novel therapies are needed for the treatment of the malignant forms of prostate cancer that recur after initial therapies, such as hormone refractory (HRPC) or castration resistant prostate cancer (CRPC). With advances in understanding of the molecular mechanisms of cancer, we have witnessed unprecedented progress in developing new forms of targeted therapy. Several targeted therapeutic agents have been developed and clinically used for the treatment of solid tumors such as breast cancer, non-small cell lung cancer, and renal cancer. Some of these reagents modulate growth factors and/or their receptors, which are abundant in cancer cells. Other reagents target the downstream signal transduction, survival pathways, and angiogenesis pathways that are abnormally activated in transformed cells or metastatic tumors. We will review current developments in this field, focusing specifically on treatments that can be applied to prostate cancers. Finally we will describe aspects of the future direction of the field with respect to discovering biomarkers to aid in identifying responsive prostate cancer patients.
Collapse
Affiliation(s)
- Weihua Fu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6082, USA
| | | | | | | |
Collapse
|
36
|
Reams RR, Kalari KR, Wang H, Odedina FT, Soliman KF, Yates C. Detecting gene-gene interactions in prostate disease in African American men. Infect Agent Cancer 2011; 6 Suppl 2:S1. [PMID: 21992608 PMCID: PMC3194179 DOI: 10.1186/1750-9378-6-s2-s1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The most common male malignancy in the United States is prostate cancer; however its rate of occurrence varies significantly among ethnic groups. In a previous cDNA microarray study on CaP tumors from African American (AA) and Caucasian (CA) patients, we identified 97 candidate genes that exhibited opposite gene expression polarity with respect to race groups; genes up-regulated in AA were simultaneously down-regulated in CA. PURPOSE The purpose of this study was to narrow the 97 member gene list, to a smaller number of genes in order to focus studies on a limited number of genes/SNPs that might explain prostate cancer disparity in African Americans. METHODS We performed genotype-phenotype, SNP and expression transcript levels correlations using HapMap Yoruba population with 85 of our 97 prostate candidate genes using SCAN database. RESULTS Findings revealed an association of SNPs surrounding ABCD3 gene with basal gene expression of RanGAP1 is important in prostate tumors in AA. Hence, to confirm our results in clinical biospecimen, we monitored expression of ABCD3 in a novel panel of African American and Caucasian prostate cancer paired cell lines. The LNCaP, C4-2B showed 2-fold increase; MDA-2PC-2B cell line, derived from AA, showed highest fold-change, 10-fold. The EGFR over expressing DU-145 WT cell line exhibited a 4-fold increase in expression relative to non transfected DU-145 prostate cell lines. Furthermore, Ingenuity Network analysis implicated our AA prostate candidate genes are involved in three network hubs, ERK, MapK and NFkB pathways. CONCLUSIONS Taken together, these findings are intriguing because other members of the ABC gene family, namely, ABCC3, ABCD1, and ABCD2 have been shown to confer chemoresistance in certain cancer types. Equally important, is the fact that activation of the MapK/ERK pathway via EGFR stimulation is vital for increased transcription of numerous cancer related genes. It is especially noteworthy that overexpression of EGFR has been widely observed in AA prostate tumors. Collectively our findings lead us to think that a novel signaling cascade, through which increased aggressiveness and chemoresistance is achieved, may explain prostate cancer health disparity in AA males and the nature of aggressive CaP tumors in general.
Collapse
Affiliation(s)
- R Renee Reams
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Weiss J, Haefeli WE. Interaction potential of the endothelin-A receptor antagonist atrasentan with drug transporters and drug-metabolising enzymes assessed in vitro. Cancer Chemother Pharmacol 2011; 68:1093-8. [PMID: 21805352 DOI: 10.1007/s00280-011-1715-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 07/15/2011] [Indexed: 01/02/2023]
Abstract
PURPOSE Atrasentan is a highly potent and selective endothelin receptor A (ET(A)) antagonist under development for the treatment of prostate cancer. Only little data exist on its interaction with drug-metabolising enzymes and drug transporters possibly influencing its safety and effectiveness. Our study evaluated whether atrasentan can induce the expression of relevant human drug transporters and cytochrome P450 isozymes (CYPs), whether it retains its efficiency in multidrug resistant cell lines, and whether it inhibits P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP). METHODS Induction of transporters and enzymes was quantified at the mRNA level by real-time RT-PCR in LS180 cells and for P-gp also at the protein level by Western blot. P-gp inhibition was evaluated by calcein assay in P388/dx and L-MDR1 cells and BCRP inhibition in MDCKII-BCRP cells by pheophorbide A efflux. Substrate characteristics were evaluated by growth inhibition assays in MDCKII cells overexpressing particular ABC-transporters. RESULTS Atrasentan profoundly induced several CYPs and drug transporters (e.g. 12-fold induction of CYP3A4 at 50 μM). It was a moderate P-gp inhibitor (IC(50) in P388/dx cells = 15.1 ± 1.6 μM) and a weak BCRP inhibitor (IC(50) in MDCKII-BCRP cells = 59.8 ± 11 μM). BCRP or P-gp overexpressing cells were slightly more resistant towards antiproliferative effects of atrasentan. CONCLUSIONS Our data provide a comprehensive analysis of the induction profile of atrasentan and its interaction with P-gp and BCRP. The profound induction effects stress the need for thorough assessment of its interaction potential in vivo.
Collapse
Affiliation(s)
- Johanna Weiss
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
| | | |
Collapse
|
38
|
Levy A, Chargari C, Desrez G, Leroux S, Sneyd MJ, Mozer P, Comperat E, Feuvret L, Lang P, Lopez S, Assouline A, Hemery C, Mazeron JJ, Simon JM. Poorer outcome in Polynesian patients with prostate cancer treated with definitive conformational radiation therapy. Radiother Oncol 2011; 101:502-7. [PMID: 21723636 DOI: 10.1016/j.radonc.2011.05.075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 05/19/2011] [Accepted: 05/26/2011] [Indexed: 10/18/2022]
Abstract
PURPOSE To compare freedom from biochemical failure (FFBF) of French Polynesian (FP) and Native European (NE) prostate cancer patients after definitive conformal radiotherapy (RT). PATIENTS AND METHODS Data were reviewed from medical records of 152 consecutive patients (46 FP and 106 NE) with clinically localised prostate cancer treated with definitive RT. Neoadjuvant androgen deprivation therapy (ADT) was used in 22% of cases. Definition for biochemical failure was a rise by 2 ng/mL or more above the nadir prostate-specific antigen (PSA) level. The median follow-up was 34 months. RESULTS In comparison to NE patients, FP patients were younger (p=0.002) with a higher low-risk proportion (p=0.06). Probability of 5-year FFBF was 77% in the NE cohort and 58.0% in the FP cohort (p=0.017). Univariate analysis showed that FP ethnicity was associated with worse prognosis in high-risk tumours (p=0.004). Cox multivariate analysis showed that factors associated with FFBF were risk category (p<0.017), and FP origin (p=0.03), independently of ADT and radiation dose. CONCLUSION FP ethnicity was an independent prognostic factor for biochemical relapse after definitive conformal RT for prostate cancer.
Collapse
Affiliation(s)
- Antonin Levy
- Department of Radiation Oncology, Pitie-Salpetriere University Hospital, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Kim HS, Moreira DM, Jayachandran J, Gerber L, Bañez LL, Vollmer RT, Lark AL, Donovan MJ, Powell D, Khan FM, Freedland SJ. Prostate biopsies from black men express higher levels of aggressive disease biomarkers than prostate biopsies from white men. Prostate Cancer Prostatic Dis 2011; 14:262-5. [DOI: 10.1038/pcan.2011.18] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
40
|
Wallace TA, Martin DN, Ambs S. Interactions among genes, tumor biology and the environment in cancer health disparities: examining the evidence on a national and global scale. Carcinogenesis 2011; 32:1107-21. [PMID: 21464040 DOI: 10.1093/carcin/bgr066] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cancer incidence and mortality rates show great variations across nations and between population groups. These variations are largely explained by differences in age distribution, diet and lifestyle, access to health care, cultural barriers and exposure to carcinogens and pathogens. Cancers caused by infections are significantly more common in developing than developed countries, and they overproportionally affect immigrant populations in the USA and other countries. The global pattern of cancer is not stagnant. Instead, it is dynamic because of fluctuations in the age distribution of populations, improvements in cancer prevention and early detection in affluent countries and rapid changes in diet and lifestyle in parts of the world. For example, increased smoking rates have caused tobacco-induced cancers to rise in various Asian countries, whereas reduced smoking rates have caused these cancers to plateau or even begin to decline in Western Europe and North America. Some population groups experience a disproportionally high cancer burden. In the USA and the Caribbean, cancer incidence and mortality rates are excessively high in populations of African ancestry when compared with other population groups. The causes of this disparity are multifaceted and may include tumor biological and genetic factors and their interaction with the environment. In this review, we will discuss the magnitude and causes of global cancer health disparities and will, with a focus on African-Americans and selected cancer sites, evaluate the evidence that genetic and tumor biological factors contribute to existing cancer incidence and outcome differences among population groups in the USA.
Collapse
Affiliation(s)
- Tiffany A Wallace
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4258, USA
| | | | | |
Collapse
|
41
|
Peraldo-Neia C, Migliardi G, Mello-Grand M, Montemurro F, Segir R, Pignochino Y, Cavalloni G, Torchio B, Mosso L, Chiorino G, Aglietta M. Epidermal Growth Factor Receptor (EGFR) mutation analysis, gene expression profiling and EGFR protein expression in primary prostate cancer. BMC Cancer 2011; 11:31. [PMID: 21266046 PMCID: PMC3040720 DOI: 10.1186/1471-2407-11-31] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 01/25/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Activating mutations of the epidermal growth factor receptor (EGFR) confer sensitivity to the tyrosine kinase inhibitors (TKi), gefitinib and erlotinib. We analysed EGFR expression, EGFR mutation status and gene expression profiles of prostate cancer (PC) to supply a rationale for EGFR targeted therapies in this disease. METHODS Mutational analysis of EGFR TK domain (exons from 18 to 21) and immunohistochemistry for EGFR were performed on tumour tissues derived from radical prostatectomy from 100 PC patients. Gene expression profiling using oligo-microarrays was also carried out in 51 of the PC samples. RESULTS EGFR protein overexpression (EGFRhigh) was found in 36% of the tumour samples, and mutations were found in 13% of samples. Patients with EGFRhigh tumours experienced a significantly increased risk of biochemical relapse (hazard ratio-HR 2.52, p=0.02) compared with patients with tumours expressing low levels of EGFR (EGFRlow). Microarray analysis did not reveal any differences in gene expression between EGFRhigh and EGFRlow tumours. Conversely, in EGFRhigh tumours, we were able to identify a 79 gene signature distinguishing mutated from non-mutated tumours. Additionally, 29 genes were found to be differentially expressed between mutated/EGFRhigh (n=3) and mutated/EGFRlow tumours (n=5). Four of the down-regulated genes, U19/EAF2, ABCC4, KLK3 and ANXA3 and one of the up-regulated genes, FOXC1, are involved in PC progression. CONCLUSIONS Based on our findings, we hypothesize that accurate definition of the EGFR status could improve prognostic stratification and we suggest a possible role for EGFR-directed therapies in PC patients. Having been generated in a relatively small sample of patients, our results warrant confirmation in larger series.
Collapse
Affiliation(s)
- Caterina Peraldo-Neia
- Department of Clinical Oncology, University of Torino Medical School, Institute for Cancer Research and Treatment, Candiolo, Turin, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hsiao W, Moses KA, Goodman M, Jani AB, Rossi PJ, Master VA. Stage IV Prostate Cancer: Survival Differences in Clinical T4, Nodal and Metastatic Disease. J Urol 2010; 184:512-8. [DOI: 10.1016/j.juro.2010.04.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2009] [Indexed: 10/19/2022]
Affiliation(s)
- Wayland Hsiao
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia
| | - Kelvin A. Moses
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia
| | - Michael Goodman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Ashesh B. Jani
- Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Peter J. Rossi
- Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Viraj A. Master
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
43
|
Rose AE, Satagopan JM, Oddoux C, Zhou Q, Xu R, Olshen AB, Yu JZ, Dash A, Jean-Gilles J, Reuter V, Gerald WL, Lee P, Osman I. Copy number and gene expression differences between African American and Caucasian American prostate cancer. J Transl Med 2010; 8:70. [PMID: 20649978 PMCID: PMC2913940 DOI: 10.1186/1479-5876-8-70] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Accepted: 07/22/2010] [Indexed: 11/26/2022] Open
Abstract
Background The goal of our study was to investigate the molecular underpinnings associated with the relatively aggressive clinical behavior of prostate cancer (PCa) in African American (AA) compared to Caucasian American (CA) patients using a genome-wide approach. Methods AA and CA patients treated with radical prostatectomy (RP) were frequency matched for age at RP, Gleason grade, and tumor stage. Array-CGH (BAC SpectralChip2600) was used to identify genomic regions with significantly different DNA copy number between the groups. Gene expression profiling of the same set of tumors was also evaluated using Affymetrix HG-U133 Plus 2.0 arrays. Concordance between copy number alteration and gene expression was examined. A second aCGH analysis was performed in a larger validation cohort using an oligo-based platform (Agilent 244K). Results BAC-based array identified 27 chromosomal regions with significantly different copy number changes between the AA and CA tumors in the first cohort (Fisher's exact test, P < 0.05). Copy number alterations in these 27 regions were also significantly associated with gene expression changes. aCGH performed in a larger, independent cohort of AA and CA tumors validated 4 of the 27 (15%) most significantly altered regions from the initial analysis (3q26, 5p15-p14, 14q32, and 16p11). Functional annotation of overlapping genes within the 4 validated regions of AA/CA DNA copy number changes revealed significant enrichment of genes related to immune response. Conclusions Our data reveal molecular alterations at the level of gene expression and DNA copy number that are specific to African American and Caucasian prostate cancer and may be related to underlying differences in immune response.
Collapse
Affiliation(s)
- Amy E Rose
- Department of Urology, New York University School of Medicine, New York, New York 10016, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Yoshio Y, Ishii K, Arase S, Hori Y, Nishikawa K, Soga N, Kise H, Arima K, Sugimura Y. Effect of transforming growth factor α overexpression on urogenital organ development in mouse. Differentiation 2010; 80:82-8. [PMID: 20638776 DOI: 10.1016/j.diff.2010.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 06/25/2010] [Accepted: 06/29/2010] [Indexed: 10/19/2022]
Abstract
Transforming growth factor-α (TGFα) promotes cell proliferation by binding to the epidermal growth factor receptor (EGFR). TGFα and EGFR overexpression have been reported in various human cancers. However, whether TGFα induces cancer by itself is unknown in urogenital organs. To investigate whether TGFα overexpression induces carcinogenesis in urogenital organs, we analyzed the phenotypes of urogenital organs in male TGFα transgenic (TG) mice of the CD1 strain. Urogenital organs including the kidney, bladder, prostate, seminal vesicles, testes, and epididymis were isolated from 4- to 48-week-old TGFα TG and wild-type (WT) CD1 mice. Prostates were separated into anterior prostate (AP), dorsolateral prostate (DLP), and ventral prostate (VP). Neither tumor formation nor epithelial hyperplasia was observed in the TGFα TG mouse urogenital organs that we have investigated. Histopathologically, in prostate, we found an increased number of p63-positive basal epithelial cells in the TGFα TG mice AP and DLP. There was no morphological change in the stromal component, such as hypercellular stroma or fibrosis. However, bladder weight was greater in TGFα TG mice than that in WT mice, and distended bladders were observed macroscopically in 19 of 20 TGFα TG mice over 20 weeks of age. Ki67 labeling index was increased significantly in the TGFα TG mouse urethral epithelium, whereas neither epithelial hyperplasia nor hypertrophy was observed. In conclusion, our results suggest that TGFα overexpression in mouse urogenital organs alone may not be responsible for tumor formation and epithelial hyperplasia, but is involved in bladder outlet obstruction.
Collapse
Affiliation(s)
- Yuko Yoshio
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Molecular alterations of EGFR and PTEN in prostate cancer: association with high-grade and advanced-stage carcinomas. Mod Pathol 2010; 23:703-12. [PMID: 20208477 DOI: 10.1038/modpathol.2010.45] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Prostate cancer is the second cause of cancer-related death in men of the Western world. The potential prognostic role of the combined alterations in EGFR and PTEN in prostate cancer is not well established. It was the aim of the study to investigate this role. Prevalence of EGFR and PTEN somatic mutations, EGFR amplification and EGFR protein expression were investigated in a series of prostate adenocarcinomas, classified according to the current Gleason grading system. Mutational analysis revealed eight EGFR and three PTEN mutations in 98 (8%) and 92 (3%) prostate adenocarcinomas, respectively. The combined prevalence of EGFR-PTEN mutations was 11%. EGFR overexpression was present in 31% of adenocarcinomas, with a marginally significant difference (P=0.068) between Gleason grade < or =7 adenocarcinomas and Gleason grade > or =8 and metastatic adenocarcinomas. Four cases (4 of 31; 13%) had an EGFR gene gain due to chromosome 7 polysomy. In 35% of adenocarcinomas we found some type of EGFR-PTEN alteration, with a tendency to be associated with advanced-stage prostate adenocarcinomas (P=0.04). The IVS18+19 polymorphism was also associated with more advanced prostate adenocarcinomas. This is the first study reporting mutations of EGFR and PTEN in the same series of prostate adenocarcinomas. Protein overexpression is the most frequent EGFR abnormality. Mutations in EGFR and PTEN genes are a minor event, although prostate cancer represents the third neoplasm in which the EGFR gene mutations are more prevalent. Alterations in the EGFR-PTEN signaling pathway are present in a third of prostate adenocarcinomas, particularly affecting the more advanced cases.
Collapse
|
46
|
Antonarakis ES, Carducci MA, Eisenberger MA. Novel targeted therapeutics for metastatic castration-resistant prostate cancer. Cancer Lett 2010; 291:1-13. [PMID: 19717225 PMCID: PMC4029098 DOI: 10.1016/j.canlet.2009.08.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Revised: 08/06/2009] [Accepted: 08/10/2009] [Indexed: 12/23/2022]
Abstract
Virtually all patients that succumb to prostate cancer die of metastatic castration-resistant disease. Although docetaxel is the standard of care for these patients and is associated with a modest prolongation of survival, there is an urgent need for novel treatment strategies for metastatic prostate cancer. In the last several years, great strides have been made in our understanding of the biological and molecular mechanisms driving prostate cancer growth and progression, and this has resulted in widespread clinical testing of numerous new targeted therapies. This review discusses some of the key therapeutic agents that have emerged for the treatment of metastatic castration-resistant prostate cancer in the last 5years, with an emphasis on both molecular targets and clinical trial design. These agents include mammalian target of rapamycin (mTOR) pathway inhibitors, anti-angiogenic drugs, epidermal growth factor receptor (EGFR) inhibitors, insulin-like growth factor (IGF) pathway inhibitors, apoptosis-inducing drugs, endothelin receptor antagonists, receptor activator of nuclear factor kappaB (RANK) ligand inhibitors, vitamin D analogues, cytochrome P17 enzyme inhibitors, androgen receptor modulators, epigenetic therapies, vaccine therapies, and cytotoxic T lymphocyte-associated antigen (CTLA)-4 blocking agents.
Collapse
Affiliation(s)
- Emmanuel S Antonarakis
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231, United States.
| | | | | |
Collapse
|
47
|
Li Y, Cozzi PJ, Russell PJ. Promising tumor-associated antigens for future prostate cancer therapy. Med Res Rev 2010; 30:67-101. [PMID: 19536865 DOI: 10.1002/med.20165] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Prostate cancer (CaP) is one of the most prevalent malignant diseases among men in Western countries. There is currently no cure for metastatic castrate-resistant CaP, and median survival for these patients is about 18 months; the high mortality rate seen is associated with widespread metastases. Progression of CaP from primary to metastatic disease is associated with several molecular and genetic changes that can affect the expression of specific tumor-associated antigens (TAAs) or receptors on the cell surface. Targeting TAAs is emerging as an area of promise for controlling late-stage and recurrent CaP. Several reviews have summarized the progress made in targeting signaling pathways for CaP but will not be discussed here. We describe some important CaP TAAs. These include prostate stem-cell antigen, prostate-specific membrane antigen, MUC1, epidermal growth factor receptor, platelet-derived growth factor and its receptor, urokinase plasminogen activator and its receptor, and extracellular matrix metalloproteinase inducer. We summarize recent advancements in our understanding of their role in CaP metastasis, as well as potential therapeutic options for targeting CaP TAAs. We also discuss the origin, identification, and characterization of prostate cancer stem cells (CSCs) and the potential benefits of targeting prostate CSCs to overcome chemoresistance and CaP recurrence.
Collapse
Affiliation(s)
- Yong Li
- Cancer Care Centre, St. George Hospital, Sydney, NSW, Australia.
| | | | | |
Collapse
|
48
|
Liu F, Wang JJ, You ZY, Zhang YD, Zhao Y. Radiosensitivity of prostate cancer cells is enhanced by EGFR inhibitor C225. Urol Oncol 2010; 28:59-66. [DOI: 10.1016/j.urolonc.2008.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 07/01/2008] [Accepted: 07/02/2008] [Indexed: 10/21/2022]
|
49
|
Joensuu G, Joensuu T, Nokisalmi P, Reddy C, Isola J, Ruutu M, Kouri M, Kupelian PA, Collan J, Pesonen S, Hemminki A. A phase I/II trial of gefitinib given concurrently with radiotherapy in patients with nonmetastatic prostate cancer. Int J Radiat Oncol Biol Phys 2009; 78:42-9. [PMID: 20004525 DOI: 10.1016/j.ijrobp.2009.07.1731] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 07/20/2009] [Accepted: 07/22/2009] [Indexed: 01/24/2023]
Abstract
PURPOSE To estimate the safety and tolerability of daily administration of 250 mg of gefitinib given concurrently with three-dimensional conformal radiotherapy for patients with nonmetastatic prostate cancer. METHODS AND MATERIALS A total of 42 patients with T2-T3N0M0 tumors were treated in a nonrandomized single-center study. A prostate-specific antigen (PSA) level of <20 and a good performance status (WHO, 0-1) were required. Adjuvant or neoadjuvant hormone treatments were not allowed. A daily regimen of 250 mg of gefitinib was started 1 week before radiation therapy began and lasted for the duration of radiation therapy. A dose of 50.4 Gy (1.8 Gy/day) was administered to the tumor, prostate, and seminal vesicles, followed by a 22-Gy booster (2 Gy/day) for a total dose of 72.4 Gy. Correlative studies included analysis of epidermal growth factor receptor (EGFR), EGFRvIII, and phosphorylated EGFR in tumors and tumor necrosis factor, interleukin-1alpha (IL-1alpha), and IL-6 in serum. RESULTS Maximum tolerated dose was not reached in phase I (12 patients), and 30 additional patients were treated in phase II. Thirty (71.4%) patients completed trial medication. Dose-limiting toxicities were recorded for 16 (38.1%) patients, the most common of which was a grade 3 to 4 increase in transaminase (6 patients). After a median follow-up of 38 months, there were no deaths due to prostate cancer. The estimated PSA relapse-free survival rate at 4 years (Kaplan-Meier) was 97%, the salvage therapy-free survival rate was 91%, and the overall survival rate was 87%. These figures compared favorably with those of matched patients treated with radiation only at higher doses. CONCLUSIONS The combination of gefitinib and radiation is reasonably well tolerated and has promising activity against nonmetastatic prostate cancer.
Collapse
Affiliation(s)
- Greetta Joensuu
- Transplantation Laboratory and Haartman Institute and Finnish Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Human epidermal growth factor receptor-2 family in colorectal adenocarcinoma: correlation with survival and clinicopathological findings. Eur J Gastroenterol Hepatol 2009; 21:289-93. [PMID: 19279475 DOI: 10.1097/meg.0b013e32830b82ba] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND PURPOSE Erb-B1 (epidermal growth factor receptor, EGFR) and Erb-B2 (HER-2) are two of the best-characterized members in the EGFR pathway. In many tumor types, overexpression of these proteins is associated with enhanced malignant potential. The aim of this study was to determine the prognostic impact of EGFR and HER-2 protein expression on colorectal cancer. METHOD Immunohistochemistry was carried out in paraffin-embedded specimens of 115 colorectal carcinomas for the assessment of EGFR and HER-2 expression. Immunostaining for EGFR was graded negative, weak or strong according to extension and staining intensity. The results were correlated with traditional clinicopathologic parameters and patients' outcome. RESULTS The mean survival time was 64 (range 9-78) months in the EGFR-negative group, 166 (range 2-293) months in the group with a low EGFR expression, and 51 (range 4-71) months in the group with a high EGFR expression. The median survival time was 31 (range 2-114) months in the HER-2 negative group, and 30 (range 4-293) months in the HER-2 positive group. None of the clinicopathologic parameters or patient prognoses had statistically significant association with EGFR or HER-2 expression. CONCLUSION Conventional immunohistochemistry was unable to reveal any association between EGFR or HER-2 expression and outcome predicted by the biologic role of EGFR in tumor behavior and the established prognostic role of HER-2 in breast cancer.
Collapse
|