1
|
Doré S, Ali M, Sorin M, McDowell SAC, Desharnais L, Breton V, Yu MW, Arabzadeh A, Ryan MI, Milette S, Quail DF, Walsh LA. Exploring the prognostic significance of arm-level copy number alterations in triple-negative breast cancer. Oncogene 2024; 43:2015-2024. [PMID: 38744952 PMCID: PMC11196216 DOI: 10.1038/s41388-024-03051-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/16/2024]
Abstract
Somatic copy number alterations (SCNAs) are prevalent in cancer and play a significant role in both tumorigenesis and therapeutic resistance. While focal SCNAs have been extensively studied, the impact of larger arm-level SCNAs remains poorly understood. Here, we investigated the association between arm-level SCNAs and overall survival in triple-negative breast cancer (TNBC), an aggressive subtype of breast cancer lacking targeted therapies. We identified frequent arm-level SCNAs, including 21q gain and 7p gain, which correlated with poor overall survival in TNBC patients. Further, we identified the expression of specific genes within these SCNAs associated with survival. Notably, we found that the expression of RIPK4, a gene located on 21q, exhibited a strong correlation with poor overall survival. In functional assays, we demonstrated that targeting Ripk4 in a murine lung metastatic TNBC model significantly reduced tumor burden, improved survival, and increased CD4+ and CD8+ T cell infiltration. RIPK4 enhanced the survival of triple-negative breast cancer cells at secondary sites, thereby facilitating the formation of metastatic lesions. Our findings highlight the significance of arm-level SCNAs in breast cancer progression and identify RIPK4 as a putative driver of TNBC metastasis and immunosuppression.
Collapse
Affiliation(s)
- Samuel Doré
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Mariam Ali
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Mark Sorin
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Sheri A C McDowell
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Lysanne Desharnais
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Valérie Breton
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Miranda W Yu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Azadeh Arabzadeh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Malcolm I Ryan
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Surgery, McGill University Health Center, Montreal, QC, Canada
| | - Simon Milette
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Logan A Walsh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
| |
Collapse
|
2
|
Yacona K, Hanna MW, Niyazi S, Sharma S, Hatch P, Hernandez M, Letter H. Can COVID-19 worsen racial disparities in breast cancer screening and diagnosis? J Clin Imaging Sci 2022; 12:35. [PMID: 36128359 PMCID: PMC9479646 DOI: 10.25259/jcis_26_2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/07/2022] [Indexed: 11/29/2022] Open
Abstract
Background Health disparities among minority groups, especially African Americans, can limit their access to quality medical care and lead to disproportionate medical management and disease outcomes. The aim of this study was to compare the COVID-19-related change in mammogram volumes and cancer detection at two affiliated academic breast centers, one that serves a predominantly African American patient population and one that serves a predominantly non-African American patient population. Materials and methods For the purpose of anonymity, racial demographics were collected and the center with a higher African American patient population was designated as institution A, while the center with a higher non-African American patient population was designated as institution B. Careful selection of the two breast centers was instituted in order to limit the impact of potential confounders other than race. An Institutional Review Board (IRB) exemption was obtained and two Mammography Quality Standards Act (MQSA) reports were generated; one for March 2020 through September 2020, during the height of the COVID-19 pandemic, and one for March 2019 through September 2019 to serve as the pre-pandemic control group. The i2b2 Query Analysis Tool® was used to obtain racial demographic data and compare the percent change in screening and diagnostic mammograms, image-guided biopsies, total cancers diagnosed by imaging, and percent of minimal cancers for both institutions. Results Screening mammograms and breast cancer detection decreased in 2020 compared to 2019 at both institutions. However, the percent change from 2019 to 2020 was greater at institution A than at institution B. Percent minimal cancers, an indicator of early-stage breast cancer also decreased more drastically at institution A than at institution B. Interestingly, the total number of diagnostic exams and image-guided biopsies increased in 2020 at institution B, whereas both decreased at institution A. Conclusion The COVID-19 pandemic may lead to worsening racial disparities in breast cancer screening. In an effort to narrow future disparity, it is crucial for radiologists and other health care providers to be aware of this inequality and educate all women on the importance of obtaining routine screening mammography. More studies are needed.
Collapse
Affiliation(s)
- Kate Yacona
- Medical School, Lake Eerie College of Osteopathic Medicine, Bradenton, Florida, United States,
| | - Mariam W Hanna
- Department of Radiology, University of Florida College of Medicine, Gainesville, Florida, United States,
| | - Sara Niyazi
- Medical School, Lake Eerie College of Osteopathic Medicine, Bradenton, Florida, United States,
| | - Smita Sharma
- Department of Radiology, University of Florida College of Medicine, Jacksonville, Florida, United States,
| | - Parlyn Hatch
- Department of Radiology, University of Florida College of Medicine, Jacksonville, Florida, United States,
| | - Mauricio Hernandez
- Department of Radiology, University of Florida College of Medicine, Jacksonville, Florida, United States,
| | - Haley Letter
- Department of Radiology, Mayo Clinic, Jacksonville, Florida, United States,
| |
Collapse
|
3
|
Oncologic Anthropology: An Interdisciplinary Approach to Understanding the Association Between Genetically Defined African Ancestry and Susceptibility for Triple Negative Breast Cancer. CURRENT BREAST CANCER REPORTS 2021. [DOI: 10.1007/s12609-021-00426-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
4
|
Trinh A, Gil Del Alcazar CR, Shukla SA, Chin K, Chang YH, Thibault G, Eng J, Jovanović B, Aldaz CM, Park SY, Jeong J, Wu C, Gray J, Polyak K. Genomic Alterations during the In Situ to Invasive Ductal Breast Carcinoma Transition Shaped by the Immune System. Mol Cancer Res 2020; 19:623-635. [PMID: 33443130 DOI: 10.1158/1541-7786.mcr-20-0949] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/19/2020] [Accepted: 12/14/2020] [Indexed: 11/16/2022]
Abstract
The drivers of ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) transition are poorly understood. Here, we conducted an integrated genomic, transcriptomic, and whole-slide image analysis to evaluate changes in copy-number profiles, mutational profiles, expression, neoantigen load, and topology in 6 cases of matched pure DCIS and recurrent IDC. We demonstrate through combined copy-number and mutational analysis that recurrent IDC can be genetically related to its pure DCIS despite long latency periods and therapeutic interventions. Immune "hot" and "cold" tumors can arise as early as DCIS and are subtype-specific. Topologic analysis showed a similar degree of pan-leukocyte-tumor mixing in both DCIS and IDC but differ when assessing specific immune subpopulations such as CD4 T cells and CD68 macrophages. Tumor-specific copy-number aberrations in MHC-I presentation machinery and losses in 3p, 4q, and 5p are associated with differences in immune signaling in estrogen receptor (ER)-negative IDC. Common oncogenic hotspot mutations in genes including TP53 and PIK3CA are predicted to be neoantigens yet are paradoxically conserved during the DCIS-to-IDC transition, and are associated with differences in immune signaling. We highlight both tumor and immune-specific changes in the transition of pure DCIS to IDC, including genetic changes in tumor cells that may have a role in modulating immune function and assist in immune escape, driving the transition to IDC. IMPLICATIONS: We demonstrate that the in situ to IDC evolutionary bottleneck is shaped by both tumor and immune cells.
Collapse
Affiliation(s)
- Anne Trinh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Carlos R Gil Del Alcazar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Sachet A Shukla
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Koei Chin
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon.,Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Young Hwan Chang
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon.,Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Guillaume Thibault
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon
| | - Jennifer Eng
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon
| | - Bojana Jovanović
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - C Marcelo Aldaz
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - So Yeon Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Joon Jeong
- Department of Surgery, Gangnam Severance Hospital, Yonsei University Medical College, Seoul, Korea
| | - Catherine Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Joe Gray
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon.,Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| |
Collapse
|
5
|
Chen Y, Sadasivan SM, She R, Datta I, Taneja K, Chitale D, Gupta N, Davis MB, Newman LA, Rogers CG, Paris PL, Li J, Rybicki BA, Levin AM. Breast and prostate cancers harbor common somatic copy number alterations that consistently differ by race and are associated with survival. BMC Med Genomics 2020; 13:116. [PMID: 32819446 PMCID: PMC7441621 DOI: 10.1186/s12920-020-00765-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 08/10/2020] [Indexed: 11/26/2022] Open
Abstract
Background Pan-cancer studies of somatic copy number alterations (SCNAs) have demonstrated common SCNA patterns across cancer types, but despite demonstrable differences in aggressiveness of some cancers by race, pan-cancer SCNA variation by race has not been explored. This study investigated a) racial differences in SCNAs in both breast and prostate cancer, b) the degree to which they are shared across cancers, and c) the impact of these shared, race-differentiated SCNAs on cancer survival. Methods Utilizing data from The Cancer Genome Atlas (TCGA), SCNAs were identified using GISTIC 2.0, and in each tumor type, differences in SCNA magnitude between African Americans (AA) and European Americans (EA) were tested using linear regression. Unsupervised hierarchical clustering of the copy number of genes residing in race-differentiated SCNAs shared between tumor types was used to identify SCNA-defined patient groups, and Cox proportional hazards regression was used to test for association between those groups and overall/progression-free survival (PFS). Results We identified SCNAs that differed by race in breast (n = 58 SCNAs; permutation p < 10− 4) and prostate tumors (n = 78 SCNAs; permutation p = 0.006). Six race-differentiated SCNAs common to breast and prostate found at chromosomes 5q11.2-q14.1, 5q15-q21.1, 8q21.11-q21.13, 8q21.3-q24.3, 11q22.3, and 13q12.3-q21.3 had consistent differences by race across both tumor types, and all six were of higher magnitude in AAs, with the chromosome 8q regions being the only amplifications. Higher magnitude copy number differences in AAs were also identified at two of these race-differentiated SCNAs in two additional hormonally-driven tumor types: endometrial (8q21.3-q24.3 and 13q12.3-q21.3) and ovarian (13q12.3-q21.3) cancers. Race differentiated SCNA-defined patient groups were significantly associated with survival differences in both cancer types, and these groups also differentiated within triple negative breast cancers based on PFS. While the frequency of the SCNA-defined patient groups differed by race, their effects on survival did not. Conclusions This study identified race-differentiated SCNAs shared by two related cancers. The association of SCNA-defined patient groups with survival demonstrates the clinical significance of combinations of these race-differentiated genomic aberrations, and the higher frequency of these alterations in AA relative to EA patients may explain racial disparities in risk of aggressive breast and prostate cancer.
Collapse
Affiliation(s)
- Yalei Chen
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA.,Center for Bioinformatics, Henry Ford Health System, Detroit, MI, USA
| | - Sudha M Sadasivan
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | - Ruicong She
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA.,Center for Bioinformatics, Henry Ford Health System, Detroit, MI, USA
| | - Indrani Datta
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA.,Center for Bioinformatics, Henry Ford Health System, Detroit, MI, USA
| | - Kanika Taneja
- Department of Pathology, Henry Ford Health System, Detroit, MI, USA
| | - Dhananjay Chitale
- Department of Pathology, Henry Ford Health System, Detroit, MI, USA.,Center for the Study of Breast Cancer Subtypes, Breast Oncology Program, Department of Surgery, Weill Cornell Medical College, New York, NY, USA
| | - Nilesh Gupta
- Department of Pathology, Henry Ford Health System, Detroit, MI, USA
| | - Melissa B Davis
- Center for the Study of Breast Cancer Subtypes, Breast Oncology Program, Department of Surgery, Weill Cornell Medical College, New York, NY, USA
| | - Lisa A Newman
- Center for the Study of Breast Cancer Subtypes, Breast Oncology Program, Department of Surgery, Weill Cornell Medical College, New York, NY, USA
| | - Craig G Rogers
- Vattikuti Urologic Institute, Henry Ford Health System, Detroit, MI, USA
| | - Pamela L Paris
- Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA
| | - Jia Li
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA.,Center for Bioinformatics, Henry Ford Health System, Detroit, MI, USA
| | - Benjamin A Rybicki
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | - Albert M Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA. .,Center for Bioinformatics, Henry Ford Health System, Detroit, MI, USA.
| |
Collapse
|
6
|
Charan M, Verma AK, Hussain S, Misri S, Mishra S, Majumder S, Ramaswamy B, Ahirwar D, Ganju RK. Molecular and Cellular Factors Associated with Racial Disparity in Breast Cancer. Int J Mol Sci 2020; 21:ijms21165936. [PMID: 32824813 PMCID: PMC7460595 DOI: 10.3390/ijms21165936] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
Recent studies have demonstrated that racial differences can influence breast cancer incidence and survival rate. African American (AA) women are at two to three fold higher risk for breast cancer than other ethnic groups. AA women with aggressive breast cancers show worse prognoses and higher mortality rates relative to Caucasian (CA) women. Over the last few years, effective treatment strategies have reduced mortality from breast cancer. Unfortunately, the breast cancer mortality rate among AA women remains higher compared to their CA counterparts. The focus of this review is to underscore the racial differences and differential regulation/expression of genetic signatures in CA and AA women with breast cancer. Moreover, immune cell infiltration significantly affects the clinical outcome of breast cancer. Here, we have reviewed recent findings on immune cell recruitment in the tumor microenvironment (TME) and documented its association with breast cancer racial disparity. In addition, we have extensively discussed the role of cytokines, chemokines, and other cell signaling molecules among AA and CA breast cancer patients. Furthermore, we have also reviewed the distinct genetic and epigenetic changes in AA and CA patients. Overall, this review article encompasses various molecular and cellular factors associated with breast cancer disparity that affects mortality and clinical outcome.
Collapse
Affiliation(s)
- Manish Charan
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Ajeet K. Verma
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Shahid Hussain
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Swati Misri
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Sanjay Mishra
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Sarmila Majumder
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA; (S.M.); (B.R.)
| | - Bhuvaneswari Ramaswamy
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA; (S.M.); (B.R.)
| | - Dinesh Ahirwar
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
- Correspondence: (D.A.); (R.K.G.)
| | - Ramesh K. Ganju
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA; (S.M.); (B.R.)
- Correspondence: (D.A.); (R.K.G.)
| |
Collapse
|
7
|
The impact of sociodemographic factors and health insurance coverage in the diagnosis and clinicopathological characteristics of breast cancer in Brazil: AMAZONA III study (GBECAM 0115). Breast Cancer Res Treat 2020; 183:749-757. [PMID: 32728860 DOI: 10.1007/s10549-020-05831-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 07/21/2020] [Indexed: 01/09/2023]
Abstract
PURPOSE In Brazil, the available cancer registries are deficient in number and quality and, hence, little information is known regarding sociodemographic, clinicopathological characteristics, treatment patterns, and outcomes of breast cancer (BC) patients. We performed the AMAZONA III/ GBECAM 0115 study and in this analysis, we describe patients' characteristics at diagnosis and their association with health insurance type. METHODS This is a prospective cohort study developed in 23 sites in Brazil including women with newly diagnosed invasive BC from January 2016 to March 2018. In order to compare healthcare insurance type, we considered patients who were treated under the Brazilian public health system as publicly insured, and women who had private insurance or paid for their treatment as privately insured. RESULTS A total of 2950 patients were included in the study. Median age at diagnosis was 53.9 years; 63.1% were publicly insured. The majority of patients (68.6%) had stage II-III breast cancer and ductal carcinoma histology (80.9%). The most common breast cancer subtype was luminal A-like (48.0%) followed by luminal B-HER2 positive-like (17.0%) and triple-negative (15.6%). Luminal A was more frequent in private (53.7% vs. 44.2%, p < .0001) than public, whereas Luminal B HER2-positive (19.2% vs. 14.2%, p = 0.0012) and HER2-positive (8.8% vs. 5.1%, p = 0.0009) were more common in patients with public health system coverage. Only 34% of patients were diagnosed by screening exams. Privately insured patients were more frequently diagnosed with stage I disease when compared to publicly insured patients; publicly insured patients had more stage III (33.5% vs. 14.7%; p-value < 0.0001) disease than privately insured ones. Breast cancer was detected by symptoms more frequently in publicly than in privately insured patients (74.2% vs 25.8%, respectively; p-value < 0.0001). CONCLUSIONS Patients with public health coverage were diagnosed with symptomatic disease, later stages and more aggressive subtypes when compared to privately insured patients.
Collapse
|
8
|
Fromont C, Atzori A, Kaur D, Hashmi L, Greco G, Cabanillas A, Nguyen HV, Jones DH, Garzón M, Varela A, Stevenson B, Iacobini GP, Lenoir M, Rajesh S, Box C, Kumar J, Grant P, Novitskaya V, Morgan J, Sorrell FJ, Redondo C, Kramer A, Harris CJ, Leighton B, Vickers SP, Cheetham SC, Kenyon C, Grabowska AM, Overduin M, Berditchevski F, Weston CJ, Knapp S, Fischer PM, Butterworth S. Discovery of Highly Selective Inhibitors of Calmodulin-Dependent Kinases That Restore Insulin Sensitivity in the Diet-Induced Obesity in Vivo Mouse Model. J Med Chem 2020; 63:6784-6801. [PMID: 32433887 PMCID: PMC7445743 DOI: 10.1021/acs.jmedchem.9b01803] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
![]()
Polymorphisms
in the region of the calmodulin-dependent kinase
isoform D (CaMK1D) gene are associated with increased incidence of
diabetes, with the most common polymorphism resulting in increased
recognition by transcription factors and increased protein expression.
While reducing CaMK1D expression has a potentially beneficial effect
on glucose processing in human hepatocytes, there are no known selective
inhibitors of CaMK1 kinases that can be used to validate or translate
these findings. Here we describe the development of a series of potent,
selective, and drug-like CaMK1 inhibitors that are able to provide
significant free target cover in mouse models and are therefore useful
as in vivo tool compounds. Our results show that
a lead compound from this series improves insulin sensitivity and
glucose control in the diet-induced obesity mouse model after both
acute and chronic administration, providing the first in vivo validation of CaMK1D as a target for diabetes therapeutics.
Collapse
Affiliation(s)
- Christophe Fromont
- Centre for Biomolecular Sciences and School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Alessio Atzori
- Centre for Biomolecular Sciences and School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Divneet Kaur
- Centre for Biomolecular Sciences and School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Lubna Hashmi
- Centre for Biomolecular Sciences and School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Graziella Greco
- School of Pharmacy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, U.K
| | - Alejandro Cabanillas
- School of Pharmacy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, U.K
| | - Huy Van Nguyen
- School of Pharmacy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, U.K
| | - D Heulyn Jones
- Division of Pharmacy and Optometry, School of Health Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M13 9PL, U.K
| | - Miguel Garzón
- Division of Pharmacy and Optometry, School of Health Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M13 9PL, U.K
| | - Ana Varela
- Division of Pharmacy and Optometry, School of Health Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M13 9PL, U.K
| | - Brett Stevenson
- Sygnature Discovery, BioCity, Pennyfoot Street, Nottingham NG1 1GF, U.K
| | - Greg P Iacobini
- Sygnature Discovery, BioCity, Pennyfoot Street, Nottingham NG1 1GF, U.K
| | - Marc Lenoir
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | - Sundaresan Rajesh
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | - Clare Box
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | - Jitendra Kumar
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Paige Grant
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Vera Novitskaya
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | - Juliet Morgan
- Sygnature Discovery, BioCity, Pennyfoot Street, Nottingham NG1 1GF, U.K
| | - Fiona J Sorrell
- Structural Genomics Consortium and Target Discovery Institute, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, U.K
| | - Clara Redondo
- Structural Genomics Consortium and Target Discovery Institute, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, U.K
| | - Andreas Kramer
- Structural Genomics Consortium and Buchmann Institute for Molecular Life Sciences, Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - C John Harris
- CJH Consultants, Ford Cottage, South Weirs, Burley Road, Brockenhurst, Hants SO42 7UQ, U.K
| | - Brendan Leighton
- The Research Network, IPC 600 Discovery Park, Ramsgate Road, Sandwich CT13 9NJ, U.K
| | - Steven P Vickers
- RenaSci Limited, BioCity, Pennyfoot Street, Nottingham NG1 1GF, U.K
| | | | - Colin Kenyon
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for Molecular and Cellular Biology, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 8000, South Africa
| | - Anna M Grabowska
- Ex Vivo Cancer Pharmacology Centre of Excellence, Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Michael Overduin
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Fedor Berditchevski
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | - Chris J Weston
- Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, U.K.,NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, B15 2TT, U.K
| | - Stefan Knapp
- Structural Genomics Consortium and Buchmann Institute for Molecular Life Sciences, Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - Peter M Fischer
- Centre for Biomolecular Sciences and School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Sam Butterworth
- Division of Pharmacy and Optometry, School of Health Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M13 9PL, U.K
| |
Collapse
|
9
|
Abstract
Adolescent and young adult (AYA) patients with cancer have not attained the same improvements in overall survival as either younger children or older adults. One possible reason for this disparity may be that the AYA cancers exhibit unique biologic characteristics, resulting in differences in clinical and treatment resistance behaviors. Our current understanding of the unique biological/genomic characteristics of AYA cancers is limited. However, there has been some progress that has provided clues about the biology of AYA cancers. We here review the latest findings in the area of AYA cancer biology and discuss what is required to advance the field for the more effective treatment of this patient population.
Collapse
|
10
|
Stenehjem DD, Hahn AW, Gill DM, Albertson D, Gowrishankar B, Merriman J, Agarwal AM, Thodima V, Harrington EB, Au TH, Maughan BL, Houldsworth J, Pal SK, Agarwal N. Predictive genomic markers of response to VEGF targeted therapy in metastatic renal cell carcinoma. PLoS One 2019; 14:e0210415. [PMID: 30682039 PMCID: PMC6347137 DOI: 10.1371/journal.pone.0210415] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 12/21/2018] [Indexed: 11/18/2022] Open
Abstract
Background First-line treatment for metastatic renal cell carcinoma (mRCC) is rapidly changing. It currently includes VEGF targeted therapies (TT), multi-target tyrosine kinase inhibitors (TKIs), mTOR inhibitors, and immunotherapy. To optimize outcomes for individual patients, genomic markers of response to therapy are needed. Here, we aim to identify tumor-based genomic markers of response to VEGF TT to optimize treatment selection. Methods From an institutional database, primary tumor tissue was obtained from 79 patients with clear cell mRCC, and targeted sequencing was performed. Clinical outcomes were obtained retrospectively. Progression-free survival (PFS) on first-line VEGF TT was correlated to genomic alterations (GAs) using Kaplan-Meier methodology and Cox proportional hazard models. A composite model of significant GAs predicting PFS in the first-line setting was developed. Results Absence of VHL mutation was associated with inferior PFS on first-line VEGF TT. A trend for inferior PFS was observed with GAs in TP53 and FLT1 C/C variant. A composite model of these 3 GAs was associated with inferior PFS in a dose-dependent manner. Conclusion In mRCC, a composite model of TP53 mutation, wild type VHL, and FLT1 C/C variant strongly predicted PFS on first-line VEGF TT in a dose-dependent manner. These findings require external validation.
Collapse
Affiliation(s)
- David D. Stenehjem
- College of Pharmacy, University of Minnesota, Duluth, MN, United States of America
| | - Andrew W. Hahn
- Department of Internal Medicine, Division of Medical Oncology, University of Utah Huntsman Cancer Institute, Salt Lake City, UT, United States of America
| | - David M. Gill
- Department of Internal Medicine, Division of Medical Oncology, University of Utah Huntsman Cancer Institute, Salt Lake City, UT, United States of America
| | - Daniel Albertson
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, United States of America
| | | | - Joseph Merriman
- Department of Internal Medicine, Division of Medical Oncology, University of Utah Huntsman Cancer Institute, Salt Lake City, UT, United States of America
| | - Archana M. Agarwal
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, United States of America
| | - Venkata Thodima
- Cancer Genetics Inc., Rutherford, NJ, United States of America
| | - Erik B. Harrington
- Pharmacotherapy Outcomes Research Center (PORC), College of Pharmacy, University of Utah, Salt Lake City, UT, United States of America
| | - Trang H. Au
- Pharmacotherapy Outcomes Research Center (PORC), College of Pharmacy, University of Utah, Salt Lake City, UT, United States of America
| | - Benjamin L. Maughan
- Department of Internal Medicine, Division of Medical Oncology, University of Utah Huntsman Cancer Institute, Salt Lake City, UT, United States of America
| | - Jane Houldsworth
- Cancer Genetics Inc., Rutherford, NJ, United States of America
- Department of Pathology, Mount Sinai School of Medicine, New York City, NY, United States of America
| | - Sumanta K. Pal
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, United States of America
| | - Neeraj Agarwal
- Department of Internal Medicine, Division of Medical Oncology, University of Utah Huntsman Cancer Institute, Salt Lake City, UT, United States of America
- * E-mail:
| |
Collapse
|
11
|
Yedjou CG, Sims JN, Miele L, Noubissi F, Lowe L, Fonseca DD, Alo RA, Payton M, Tchounwou PB. Health and Racial Disparity in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1152:31-49. [PMID: 31456178 PMCID: PMC6941147 DOI: 10.1007/978-3-030-20301-6_3] [Citation(s) in RCA: 247] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Breast cancer is the most common noncutaneous malignancy and the second most lethal form of cancer among women in the United States. It currently affects more than one in ten women worldwide. The chance for a female to be diagnosed with breast cancer during her lifetime has significantly increased from 1 in 11 women in 1975 to 1 in 8 women (Altekruse, SEER Cancer Statistics Review, 1975-2007. National Cancer Institute, Bethesda, 2010). This chance for a female of being diagnosed with cancer generally increases with age (Howlader et al, SEER Cancer Statistics Review, 1975-2010. National Cancer Institute, Bethesda, 2013). Fortunately, the mortality rate from breast cancer has decreased in recent years due to increased emphasis on early detection and more effective treatments in the White population. Although the mortality rates have declined in some ethnic populations, the overall cancer incidence among African American and Hispanic population has continued to grow. The goal of the work presented in this book chapter is to highlight similarities and differences in breast cancer morbidity and mortality rates among non-Hispanic white and non-Hispanic black populations. This book chapter also provides an overview of breast cancer, racial/ethnic disparities in breast cancer, breast cancer incidence and mortality rate linked to hereditary, major risk factors of breast cancer among minority population, breast cancer treatment, and health disparity. A considerable amount of breast cancer treatment research have been conducted, but with limited success for African Americans compared to other ethnic groups. Therefore, new strategies and approaches are needed to promote breast cancer prevention, improve survival rates, reduce breast cancer mortality, and ultimately improve the health outcomes of racial/ethnic minorities. In addition, it is vital that leaders and medical professionals from minority population groups be represented in decision-making in research so that racial disparity in breast cancer can be well-studied, fully addressed, and ultimately eliminated in breast cancer.
Collapse
Affiliation(s)
- Clement G Yedjou
- Natural Chemotherapeutics Research Laboratory, NIH/NIMHD RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, Jackson, MS, USA.
| | - Jennifer N Sims
- Department of Epidemiology and Biostatistics, College of Public Service, Jackson State University, Jackson Medical Mall - Thad Cochran Center, Jackson, MS, USA
| | - Lucio Miele
- LSU Health Sciences Center, School of Medicine, Department of Genetics, New Orleans, LA, USA
| | - Felicite Noubissi
- Natural Chemotherapeutics Research Laboratory, NIH/NIMHD RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, Jackson, MS, USA
| | - Leroy Lowe
- Getting to Know Cancer (NGO), Truro, NS, Canada
| | - Duber D Fonseca
- Natural Chemotherapeutics Research Laboratory, NIH/NIMHD RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, Jackson, MS, USA
| | - Richard A Alo
- Natural Chemotherapeutics Research Laboratory, NIH/NIMHD RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, Jackson, MS, USA
| | - Marinelle Payton
- Department of Epidemiology and Biostatistics, College of Public Service, Jackson State University, Jackson Medical Mall - Thad Cochran Center, Jackson, MS, USA
| | - Paul B Tchounwou
- Natural Chemotherapeutics Research Laboratory, NIH/NIMHD RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, Jackson, MS, USA
| |
Collapse
|
12
|
Lupicki K, Elifio-Esposito S, Fonseca AS, Weber SH, Sugita B, Langa BC, Pereira SRF, Govender D, Panieri E, Hiss DC, Abdul-Rasool S, Cavalli LR. Patterns of copy number alterations in primary breast tumors of South African patients and their impact on functional cellular pathways. Int J Oncol 2018; 53:2745-2757. [PMID: 30320392 DOI: 10.3892/ijo.2018.4589] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/24/2018] [Indexed: 11/05/2022] Open
Abstract
Breast cancer is the most common and the leading cause of female mortality among South African (SA) women. Several non‑biological and biological risk factors may be attributed to their observed high mortality rate; however, the molecular profiles associated with their breast tumors are poorly characterized. The present study examined the patterns of genome-wide copy number alterations (CNAs) and their potential impact on functional cellular pathways targeted by cancer driver genes in patients with breast cancer from the Western Cape region of SA. Array-comparative genomic hybridization analysis, performed in 28 cases of invasive breast cancer, revealed a mean number of 8.68±6.18 CNAs per case, affecting primarily the Xp22.3 and 6p21-p25 cytobands (57.14% of the cases), followed by 19p13.3-p13.11 (35.7%), 2p25.3-p24.3, 4p16.3-p15.3, 8q11.1-q24.3 and 16 p13.3-p11.2 (32.14%). Functional enrichment analysis of genes and microRNA targets mapped in these affected cytobands revealed critical cancer-associated pathways, including fatty acid biosynthesis and metabolism, extracellular matrix-receptor interaction, hippo and tumor protein p53 signaling pathways, which are regulated by known cancer genes, including CCND1, CDKN1A, MAPK1, MDM2, TP53 and SMAD2. An inverse correlation was observed among the number of CNAs and tumor size and grade; CNAs on the 4p and 6p cytobands were also inversely correlated with tumor grade. No association was observed in the number of CNAs and/or the affected cytobands and the different ethnic groups of the SA patients, indicating that their tumor genome is affected by CNAs, irrespectively of their genetic descent. Additional genomic tumor profiling in SA and other Sub-Saharan African patients with breast cancer is required to determine the associations of the CNAs observed with prognosis and clinical outcome.
Collapse
Affiliation(s)
- Kamil Lupicki
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC 20007, USA
| | - Selene Elifio-Esposito
- Department of Health Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná 80215-901, Brazil
| | - Aline S Fonseca
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC 20007, USA
| | - Saulo H Weber
- Department of Biotechnology, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná 80215-901, Brazil
| | - Bruna Sugita
- Department of Genetics, Federal University of Parana, Curitiba, Paraná 80060-000, Brazil
| | - Bridget C Langa
- Department of Medical Biosciences, University of the Western Cape, Bellville, Western Cape 7535, South Africa
| | - Silma R F Pereira
- Department of Biology, Federal University of Maranhão, São Luis, Maranhão 65080-805, Brazil
| | - Dhirendra Govender
- Division of Anatomical Pathology, University of Cape Town, National Health Laboratory Service, Groote Schuur Hospital, Cape Town 7700, South Africa
| | - Eugene Panieri
- Department of Surgical Oncology, Groote Schuur Hospital, Cape Town 7700, South Africa
| | - Donavon C Hiss
- Department of Medical Biosciences, University of the Western Cape, Bellville, Western Cape 7535, South Africa
| | - Sahar Abdul-Rasool
- Department of Medical Biosciences, University of the Western Cape, Bellville, Western Cape 7535, South Africa
| | - Luciane R Cavalli
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC 20007, USA
| |
Collapse
|
13
|
Yuan J, Hu Z, Mahal BA, Zhao SD, Kensler KH, Pi J, Hu X, Zhang Y, Wang Y, Jiang J, Li C, Zhong X, Montone KT, Guan G, Tanyi JL, Fan Y, Xu X, Morgan MA, Long M, Zhang Y, Zhang R, Sood AK, Rebbeck TR, Dang CV, Zhang L. Integrated Analysis of Genetic Ancestry and Genomic Alterations across Cancers. Cancer Cell 2018; 34:549-560.e9. [PMID: 30300578 PMCID: PMC6348897 DOI: 10.1016/j.ccell.2018.08.019] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 06/08/2018] [Accepted: 08/29/2018] [Indexed: 12/22/2022]
Abstract
Disparities in cancer care have been a long-standing challenge. We estimated the genetic ancestry of The Cancer Genome Atlas patients, and performed a pan-cancer analysis on the influence of genetic ancestry on genomic alterations. Compared with European Americans, African Americans (AA) with breast, head and neck, and endometrial cancers exhibit a higher level of chromosomal instability, while a lower level of chromosomal instability was observed in AAs with kidney cancers. The frequencies of TP53 mutations and amplification of CCNE1 were increased in AAs in the cancer types showing higher levels of chromosomal instability. We observed lower frequencies of genomic alterations affecting genes in the PI3K pathway in AA patients across cancers. Our result provides insight into genomic contribution to cancer disparities.
Collapse
Affiliation(s)
- Jiao Yuan
- Center for Research on Reproduction & Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhongyi Hu
- Center for Research on Reproduction & Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brandon A Mahal
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sihai D Zhao
- Department of Statistics, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA
| | - Kevin H Kensler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - Jingjiang Pi
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiaowen Hu
- Center for Research on Reproduction & Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Youyou Zhang
- Center for Research on Reproduction & Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yueying Wang
- Center for Research on Reproduction & Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Junjie Jiang
- Center for Research on Reproduction & Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chunsheng Li
- Center for Research on Reproduction & Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaomin Zhong
- Center for Stem Cell Biology and Tissue Engineering, Department of Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Kathleen T Montone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guoqiang Guan
- Department of Orthodontics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Janos L Tanyi
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark A Morgan
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Meixiao Long
- Department of Internal Medicine, Division of Hematology, Ohio State University, Columbus, OH 43210, USA
| | - Yuzhen Zhang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | | | - Anil K Sood
- Center for RNA Interference and Non-coding RNA, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77584, USA
| | - Timothy R Rebbeck
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - Chi V Dang
- Wistar Institute, Philadelphia, PA 19104, USA; Ludwig Institute for Cancer Research, New York City, NY 10017, USA
| | - Lin Zhang
- Center for Research on Reproduction & Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
14
|
Naab TJ, Gautam A, Ricks-Santi L, Esnakula AK, Kanaan YM, DeWitty RL, Asgedom G, Makambi KH, Abawi M, Blancato JK. MYC amplification in subtypes of breast cancers in African American women. BMC Cancer 2018. [PMID: 29523126 PMCID: PMC5845301 DOI: 10.1186/s12885-018-4171-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background MYC overexpression is associated with poor prognosis in breast tumors (BCa). The objective of this study was to determine the prevalence of MYC amplification and associated markers in BCa tumors from African American (AA) women and determine the associations between MYC amplification and clinico-pathological characteristics. Methods We analyzed 70 cases of well characterized archival breast ductal carcinoma specimens from AA women for MYC oncogene amplification. Utilizing immune histochemical analysis estrogen receptor (ER), progesterone receptor (PR), and (HER2/neu), were assessed. Cases were Luminal A (ER or PR+, Ki-67 < 14%), Luminal B (ER or PR+, Ki-67 = > 14% or ER or PR+ HER2+), HER2 (ER-, PR-, HER2+), and Triple Negative (ER-, PR-, HER2-) with basal-like phenotype. The relationship between MYC amplification and prognostic clinico-pathological characteristics was determined using chi square and logistic regression modeling. Results Sixty-five (97%) of the tumors showed MYC gene amplification (MYC: CEP8 > 1). Statistically significant associations were found between MYC amplification and HER2-amplified BCa, and Luminal B subtypes of BCa (p < 0.0001), stage (p < 0.001), metastasis (p < 0.001), and positive lymph node status (p = 0.039). MYC amplification was associated with HER2 status (p = 0.01) and tumor size (p = 0.01). High MYC amplification was seen in grade III carcinomas (MYC: CEP8 = 2.42), pre-menopausal women (MYC: CEP8 = 2.49), PR-negative status (MYC: CEP8 = 2.42), and ER-positive status (MYC: CEP8 = 2.4). Conclusions HER2 positive BCas in AA women are likely to exhibit MYC amplification. High amplification ratios suggest that MYC drives HER2 amplification, especially in HER2 positive, Luminal B, and subtypes of BCa.
Collapse
Affiliation(s)
- Tammey J Naab
- Department of Pathology, Howard University College of Medicine, Howard University Hospital, 2041 Georgia Avenue Rm. 1M-06, Washington DC, NW, 20060, USA
| | - Anita Gautam
- Department of Oncology, University of Massachusetts Medical School, 373 Plantation street Suite# 318, Worcester, MA, 01581, England
| | - Luisel Ricks-Santi
- Cancer Research Center, Department of Biological Sciences, Hampton University, 100 E. Queen Street, Hampton, VA, 23668, USA
| | - Ashwini K Esnakula
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, P.O. Box 100275, 1600 SW Archer Road, Gainesville, FL, 32610-0275, USA
| | - Yasmine M Kanaan
- Department of Microbiology, Howard University College of Medicine, 2041 Georgia Avenue Rm. 1M-06, Washington DC, NW, 20060, USA
| | - Robert L DeWitty
- Department of Surgery, Howard University Hospital, 2041 Georgia Avenue, Washington DC, NW, 20060, USA
| | - Girmay Asgedom
- Department of Medicine, Howard University Hospital, 2041 Georgia Avenue, Washington DC, NW, 20060, USA
| | - Khepher H Makambi
- Department of Biostatistics, Bioinformatics, and Biomathematics, Lombardi Comprehensive Cancer Center, Georgetown University, 4000 Reservoir Road, Washington, DC, NW, 20057, USA
| | - Massih Abawi
- Inherited Cancer Program, GeneDx, 207 Perry Pkwy, Gaithersburg, MD, 20877, USA
| | - Jan K Blancato
- Department of Oncology, Lombardi Comprehensive Cancer Centre, Georgetown University Medical Centre, 3800 Reservoir Road, Washington DC, NW, 20007, USA.
| |
Collapse
|
15
|
Sugita B, Gill M, Mahajan A, Duttargi A, Kirolikar S, Almeida R, Regis K, Oluwasanmi OL, Marchi F, Marian C, Makambi K, Kallakury B, Sheahan L, Cavalli IJ, Ribeiro EM, Madhavan S, Boca S, Gusev Y, Cavalli LR. Differentially expressed miRNAs in triple negative breast cancer between African-American and non-Hispanic white women. Oncotarget 2018; 7:79274-79291. [PMID: 27813494 PMCID: PMC5346713 DOI: 10.18632/oncotarget.13024] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 10/25/2016] [Indexed: 01/09/2023] Open
Abstract
Triple Negative Breast Cancer (TNBC), a clinically aggressive subtype of breast cancer, disproportionately affects African American (AA) women when compared to non-Hispanic Whites (NHW). MiRNAs(miRNAs) play a critical role in these tumors, through the regulation of cancer driver genes. In this study, our goal was to characterize and compare the patterns of miRNA expression in TNBC of AA (n = 27) and NHW women (n = 30). A total of 256 miRNAs were differentially expressed between these groups, and distinct from the ones observed in their respective non-TNBC subtypes. Fifty-five of these miRNAs were mapped in cytobands carrying copy number alterations (CNAs); 26 of them presented expression levels concordant with the observed CNAs. Receiving operating characteristic (ROC) analysis showed a good power (AUC ≥ 0.80; 95% CI) for over 65% of the individual miRNAs and a high combined power with superior sensitivity and specificity (AUC = 0.88 (0.78−0.99); 95% CI) of the 26 miRNA panel in discriminating TNBC between these populations. Subsequent miRNA target analysis revealed their involvement in the interconnected PI3K/AKT, MAPK and insulin signaling pathways. Additionally, three miRNAs of this panel were associated with early age at diagnosis. Altogether, these findings indicated that there are different patterns of miRNA expression between TNBC of AA and NHW women and that their mapping in genomic regions with high levels of CNAs is not merely physical, but biologically relevant to the TNBC phenotype. Once validated in distinct cohorts of AA women, this panel can potentially represent their intrinsic TNBC genome signature.
Collapse
Affiliation(s)
- Bruna Sugita
- Department of Genetics, Federal University of Paraná, Curitiba, PR, Brazil
| | - Mandeep Gill
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Akanskha Mahajan
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Anju Duttargi
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Saurabh Kirolikar
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Rodrigo Almeida
- Department of Genetics, Federal University of Paraná, Curitiba, PR, Brazil
| | - Kenny Regis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Olusayo L Oluwasanmi
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Fabio Marchi
- International Research Center-CIPE, A. C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Catalin Marian
- The Ohio State University Comprehensive Cancer Center, Division of Cancer Prevention and Control, College of Medicine, The Ohio State University, Columbus, Ohio.,The University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| | - Kepher Makambi
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Departments of Biostatistics, Bioinformatics, and Biomathematics, Georgetown University, Washington, DC USA
| | - Bhaskar Kallakury
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
| | - Laura Sheahan
- Innovation Center for Biomedical Informatics, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Iglenir J Cavalli
- Department of Genetics, Federal University of Paraná, Curitiba, PR, Brazil
| | - Enilze M Ribeiro
- Department of Genetics, Federal University of Paraná, Curitiba, PR, Brazil
| | - Subha Madhavan
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Innovation Center for Biomedical Informatics, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Simina Boca
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Innovation Center for Biomedical Informatics, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Yuriy Gusev
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Innovation Center for Biomedical Informatics, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Luciane R Cavalli
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
16
|
Deshmukh SK, Srivastava SK, Tyagi N, Ahmad A, Singh AP, Ghadhban AAL, Dyess DL, Carter JE, Dugger K, Singh S. Emerging evidence for the role of differential tumor microenvironment in breast cancer racial disparity: a closer look at the surroundings. Carcinogenesis 2017; 38:757-765. [PMID: 28430867 DOI: 10.1093/carcin/bgx037] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/28/2017] [Indexed: 12/24/2022] Open
Abstract
Although increased awareness leading to early detection and prevention, as well as advancements in treatment strategies, have resulted in superior clinical outcomes, African American women with breast cancer continue to have greater mortality rates, compared to Caucasian American counterparts. Moreover, African American women are more likely to have breast cancer at a younger age and be diagnosed with aggressive tumor sub-types. Such racial disparities can be attributed to socioeconomic differences, but it is increasingly being recognized that these disparities may indeed be due to certain genetic and other non-genetic biological differences. Tumor microenvironment, which provides a favorable niche for the growth of tumor cells, is comprised of several types of stromal cells and the various proteins secreted as a consequence of bi-directional tumor-stromal cross-talk. Emerging evidence suggests inherent biological differences in the tumor microenvironment of breast cancer patients from different racial backgrounds. Tumor microenvironment components, affected by the genetic make-up of the tumor cells as well as other non-tumor-associated factors, may also render patients more susceptible to the development of aggressive tumors and faster progression of disease resulting in early onset, thus adversely affecting patients' survival. This review provides an overview of breast cancer racial disparity and discusses the existence of race-associated differential tumor microenvironment and its underlying genetic and non-genetic causal factors. A better understanding of these aspects would help further research on effective cancer management and improved approaches for reducing the racial disparities gaps in breast cancer patients.
Collapse
Affiliation(s)
- Sachin Kumar Deshmukh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Sanjeev K Srivastava
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.,Division of Cell Biology and Genetics, Tatva Biosciences, Coastal Innovation Hub, 600 Clinic Drive, 3rd Floor, Mobile, AL 36688, USA
| | - Nikhil Tyagi
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Aamir Ahmad
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Ajay P Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Ahmed A L Ghadhban
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Donna L Dyess
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - James E Carter
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL 36617, USA
| | - Kari Dugger
- Department of Clinical and Diagnostic Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Seema Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| |
Collapse
|
17
|
Chandler MR, Keene KS, Tuomela JM, Forero-Torres A, Desmond R, Vuopala KS, Harris KW, Merner ND, Selander KS. Lower frequency of TLR9 variant associated with protection from breast cancer among African Americans. PLoS One 2017; 12:e0183832. [PMID: 28886076 PMCID: PMC5590816 DOI: 10.1371/journal.pone.0183832] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/11/2017] [Indexed: 12/11/2022] Open
Abstract
Introduction Toll-like receptor 9 (TLR9) is an innate immune system DNA-receptor that regulates tumor invasion and immunity in vitro. Low tumor TLR9 expression has been associated with poor survival in Caucasian patients with triple negative breast cancer (TNBC). African American (AA) patients with TNBC have worse prognosis than Caucasians but whether this is due to differences in tumor biology remains controversial. We studied the prognostic significance of tumor Toll like receptor-9 (TLR9) protein expression among African American (AA) triple negative breast cancer (TNBC) patients. Germline TLR9 variants in European Americans (EAs) and AAs were investigated, to determine their contribution to AA breast cancer risk. Methods TLR9 expression was studied with immunohistochemistry in archival tumors. Exome Variant Server and The Cancer Genome Atlas were used to determine the genetic variation in the general EA and AA populations, and AA breast cancer cases. Minor allele frequencies (MAFs) were compared between EAs (n = 4300), AAs (n = 2203), and/or AA breast cancer cases (n = 131). Results Thirty-two TLR9 variants had a statistically significant MAF difference between general EAs and AAs. Twenty-one of them affect a CpG site. Rs352140, a variant previously associated with protection from breast cancer, is more common in EAs than AAs (p = 2.20E-16). EAs had more synonymous alleles, while AAs had more rare coding alleles. Similar analyses comparing AA breast cancer cases with AA controls did not reveal any variant class differences; however, three previously unreported TLR9 variants were associated with late onset breast cancer. Although not statistically significant, rs352140 was observed less frequently in AA cases compared to controls. Tumor TLR9 protein expression was not associated with prognosis. Conclusions Tumor TLR9 expression is not associated with prognosis in AA TNBC. Significant differences were detected in TLR9 variant MAFs between EAs and AAs. They may affect TLR9 expression and function. Rs352140, which may protect from breast cancer, is 1.6 X more common among EAs. These findings call for a detailed analysis of the contribution of TLR9 to breast cancer pathophysiology and health disparities.
Collapse
Affiliation(s)
- Madison R. Chandler
- Harrison School of Pharmacy, Auburn University, Auburn, AL, United States of America
| | - Kimberly S. Keene
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Johanna M. Tuomela
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Andres Forero-Torres
- Department of Medicine, Division of Hematology & Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Renee Desmond
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Department of Medicine, Division of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Katri S. Vuopala
- Department of Pathology, Lapland Central Hospital, Rovaniemi, Finland
| | - Kevin W. Harris
- Department of Medicine, Division of Hematology & Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Birmingham Veterans Affairs Medical Center, Birmingham, AL, United States of America
| | - Nancy D. Merner
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
| | - Katri S. Selander
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Department of Pathology, Lapland Central Hospital, Rovaniemi, Finland
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL, United States of America
- * E-mail:
| |
Collapse
|
18
|
Yedjou CG, Tchounwou PB, Payton M, Miele L, Fonseca DD, Lowe L, Alo RA. Assessing the Racial and Ethnic Disparities in Breast Cancer Mortality in the United States. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:E486. [PMID: 28475137 PMCID: PMC5451937 DOI: 10.3390/ijerph14050486] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 04/22/2017] [Accepted: 04/26/2017] [Indexed: 01/01/2023]
Abstract
Breast cancer is the second leading cause of cancer related deaths among women aged 40-55 in the United States and currently affects more than one in ten women worldwide. It is also one of the most diagnosed cancers in women both in wealthy and poor countries. Fortunately, the mortality rate from breast cancer has decreased in recent years due to increased emphasis on early detection and more effective treatments in White population. Although the mortality rates have declined in some ethnic populations, the overall cancer incidence among African American and Hispanic populations has continued to grow. The goal of the present review article was to highlight similarities and differences in breast cancer morbidity and mortality rates primarily among African American women compared to White women in the United States. To reach our goal, we conducted a search of articles in journals with a primary focus on minority health, and authors who had published articles on racial/ethnic disparity related to breast cancer patients. A systematic search of original research was conducted using MEDLINE, PUBMED and Google Scholar databases. We found that racial/ethnic disparities in breast cancer may be attributed to a large number of clinical and non-clinical risk factors including lack of medical coverage, barriers to early detection and screening, more advanced stage of disease at diagnosis among minorities, and unequal access to improvements in cancer treatment. Many African American women have frequent unknown or unstaged breast cancers than White women. These risk factors may explain the differences in breast cancer treatment and survival rate between African American women and White women. New strategies and approaches are needed to promote breast cancer prevention, improve survival rate, reduce breast cancer mortality, and ultimately improve the health outcomes of racial/ethnic minorities.
Collapse
Affiliation(s)
- Clement G Yedjou
- Natural Chemotherapeutics Research Laboratory, Research Centers in Minority Institutio (RCMI)-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA.
| | - Paul B Tchounwou
- Natural Chemotherapeutics Research Laboratory, Research Centers in Minority Institutio (RCMI)-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA.
| | - Marinelle Payton
- Center of Excellence in Minority Health and Health Disparities, School of Public Health, Jackson State University, Jackson Medical Mall-Thad Cochran Center, 350 West Woodrow Wilson Avenue, Jackson, MS 39213, USA.
| | - Lucio Miele
- Department of Genetics, LSU Health Sciences Center, School of Medicine, 533 Bolivar Street, Room 657, New Orleans, LA 70112, USA.
| | - Duber D Fonseca
- Natural Chemotherapeutics Research Laboratory, Research Centers in Minority Institutio (RCMI)-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA.
| | - Leroy Lowe
- Lancaster Environment Centre, Lancaster University, Bailrigg, Lancaster LA1 4YW, UK.
| | - Richard A Alo
- Department of Civil and Environmental Engineering, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA.
| |
Collapse
|
19
|
Milioli HH, Tishchenko I, Riveros C, Berretta R, Moscato P. Basal-like breast cancer: molecular profiles, clinical features and survival outcomes. BMC Med Genomics 2017; 10:19. [PMID: 28351365 PMCID: PMC5370447 DOI: 10.1186/s12920-017-0250-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 03/03/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Basal-like constitutes an important molecular subtype of breast cancer characterised by an aggressive behaviour and a limited therapy response. The outcome of patients within this subtype is, however, divergent. Some individuals show an increased risk of dying in the first five years, and others a long-term survival of over ten years after the diagnosis. In this study, we aim at identifying markers associated with basal-like patients' survival and characterising subgroups with distinct disease outcome. METHODS We explored the genomic and transcriptomic profiles of 351 basal-like samples from the METABRIC and ROCK data sets. Two selection methods, labelled Differential and Survival filters, were employed to determine genes/probes that are differentially expressed in tumour and control samples, and are associated with overall survival. These probes were further used to define molecular subgroups, which vary at the microRNA level and in DNA copy number. RESULTS We identified the expression signature of 80 probes that distinguishes between two basal-like subgroups with distinct clinical features and survival outcomes. Genes included in this list have been mainly linked to cancer immune response, epithelial-mesenchymal transition and cell cycle. In particular, high levels of CXCR6, HCST, C3AR1 and FPR3 were found in Basal I; whereas HJURP, RRP12 and DNMT3B appeared over-expressed in Basal II. These genes exhibited the highest betweenness centrality and node degree values and play a key role in the basal-like breast cancer differentiation. Further molecular analysis revealed 17 miRNAs correlated to the subgroups, including hsa-miR-342-5p, -150, -155, -200c and -17. Additionally, increased percentages of gains/amplifications were detected on chromosomes 1q, 3q, 8q, 10p and 17q, and losses/deletions on 4q, 5q, 8p and X, associated with reduced survival. CONCLUSIONS The proposed signature supports the existence of at least two subgroups of basal-like breast cancers with distinct disease outcome. The identification of patients at a low risk may impact the clinical decisions-making by reducing the prescription of high-dose chemotherapy and, consequently, avoiding adverse effects. The recognition of other aggressive features within this subtype may be also critical for improving individual care and for delineating more effective therapies for patients at high risk.
Collapse
Affiliation(s)
- Heloisa H. Milioli
- Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, New Lambton Heights, 2305 Australia
- School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, 2308 Australia
| | - Inna Tishchenko
- Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, New Lambton Heights, 2305 Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, University Drive, Callaghan, 2308 Australia
| | - Carlos Riveros
- CReDITSS Unit, Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, New Lambton Heights, 2305 Australia
| | - Regina Berretta
- Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, New Lambton Heights, 2305 Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, University Drive, Callaghan, 2308 Australia
| | - Pablo Moscato
- Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, New Lambton Heights, 2305 Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, University Drive, Callaghan, 2308 Australia
| |
Collapse
|
20
|
Özdemir BC, Dotto GP. Racial Differences in Cancer Susceptibility and Survival: More Than the Color of the Skin? Trends Cancer 2017; 3:181-197. [PMID: 28718431 DOI: 10.1016/j.trecan.2017.02.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/03/2017] [Accepted: 02/06/2017] [Indexed: 12/14/2022]
Abstract
Epidemiological studies point to race as a determining factor in cancer susceptibility. In US registries recording cancer incidence and survival by race (distinguishing 'black versus white'), individuals of African ancestry have a globally increased risk of malignancies compared with Caucasians and Asian Americans. Differences in socioeconomic status and health-care access play a key role. However, the lesser disease susceptibility of Hispanic populations with comparable lifestyles and socioeconomic status as African Americans (Hispanic paradox) points to the concomitant importance of genetic determinants. Here, we overview the molecular basis of racial disparity in cancer susceptibility ranging from genetic polymorphisms and cancer-driver gene mutations to obesity, chronic inflammation, and immune responses. We discuss implications for race-adapted cancer screening programs and clinical trials to reduce disparities in cancer burden.
Collapse
Affiliation(s)
- Berna C Özdemir
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Rue du Bugnon 46, 1011 Lausanne, Switzerland
| | - Gian-Paolo Dotto
- Department of Biochemistry, University of Lausanne, Chemin des Boveresses 155, 1066 Épalinges, Switzerland; Harvard Dermatology Department and Cutaneous Biology Research Center, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02129, USA.
| |
Collapse
|
21
|
Tricoli JV, Bleyer A, Anninga J, Barr R. The Biology of AYA Cancers. CANCER IN ADOLESCENTS AND YOUNG ADULTS 2017. [DOI: 10.1007/978-3-319-33679-4_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
22
|
Tracking Cancer Genetic Evolution using OncoTrack. Sci Rep 2016; 6:29647. [PMID: 27412732 PMCID: PMC4944131 DOI: 10.1038/srep29647] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 06/20/2016] [Indexed: 02/07/2023] Open
Abstract
It is difficult for existing methods to quantify, and track the constant evolution of cancers due to high heterogeneity of mutations. However, structural variations associated with nucleotide number changes show repeatable patterns in localized regions of the genome. Here we introduce SPKMG, which generalizes nucleotide number based properties of genes, in statistical terms, at the genome-wide scale. It is measured from the normalized amount of aligned NGS reads in exonic regions of a gene. SPKMG values are calculated within OncoTrack. SPKMG values being continuous numeric variables provide a statistical metric to track DNA level changes. We show that SPKMG measures of cancer DNA show a normative pattern at the genome-wide scale. The analysis leads to the discovery of core cancer genes and also provides novel dynamic insights into the stage of cancer, including cancer development, progression, and metastasis. This technique will allow exome data to also be used for quantitative LOH/CNV analysis for tracking tumour progression and evolution with a higher efficiency.
Collapse
|
23
|
Li G, Wu X, Qian W, Cai H, Sun X, Zhang W, Tan S, Wu Z, Qian P, Ding K, Lu X, Zhang X, Yan H, Song H, Guang S, Wu Q, Lobie PE, Shan G, Zhu T. CCAR1 5' UTR as a natural miRancer of miR-1254 overrides tamoxifen resistance. Cell Res 2016; 26:655-73. [PMID: 27002217 PMCID: PMC4897177 DOI: 10.1038/cr.2016.32] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 02/03/2016] [Accepted: 02/03/2016] [Indexed: 01/05/2023] Open
Abstract
MicroRNAs (miRNAs) typically bind to unstructured miRNA-binding sites in target RNAs, leading to a mutual repression of expression. Here, we report that miR-1254 interacts with structured elements in cell cycle and apoptosis regulator 1 (CCAR1) 5′ untranslated region (UTR) and this interaction enhances the stability of both molecules. miR-1254 can also act as a repressor when binding to unstructured sites in its targets. Interestingly, structured miR-1254-targeting sites act as both a functional RNA motif-sensing unit, and an independent RNA functional unit that enhances miR-1254 expression. Artificially designed miRNA enhancers, termed “miRancers”, can stabilize and enhance the activity of miRNAs of interest. We further demonstrate that CCAR1 5′ UTR as a natural miRancer of endogenous miR-1254 re-sensitizes tamoxifen-resistant breast cancer cells to tamoxifen. Thus, our study presents a novel model of miRNA function, wherein highly structured miRancer-like motif-containing RNA fragments or miRancer molecules specifically interact with miRNAs, leading to reciprocal stabilization.
Collapse
Affiliation(s)
- Gaopeng Li
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xiaoli Wu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Wenchang Qian
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Huayong Cai
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xinbao Sun
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Weijie Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Sheng Tan
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Zhengsheng Wu
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230027, China
| | - Pengxu Qian
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA
| | - Keshuo Ding
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230027, China
| | - Xuefei Lu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Xiao Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Hong Yan
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230027, China
| | - Haifeng Song
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Shouhong Guang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Qingfa Wu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Peter E Lobie
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore 117599, Singapore
| | - Ge Shan
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Tao Zhu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| |
Collapse
|
24
|
Tricoli JV, Blair DG, Anders CK, Bleyer WA, Boardman LA, Khan J, Kummar S, Hayes-Lattin B, Hunger SP, Merchant M, Seibel NL, Thurin M, Willman CL. Biologic and clinical characteristics of adolescent and young adult cancers: Acute lymphoblastic leukemia, colorectal cancer, breast cancer, melanoma, and sarcoma. Cancer 2016; 122:1017-28. [PMID: 26849082 DOI: 10.1002/cncr.29871] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/03/2015] [Accepted: 12/04/2015] [Indexed: 01/20/2023]
Abstract
Adolescent and young adult (AYA) patients with cancer have not attained the same improvements in overall survival as either younger children or older adults. One possible reason for this disparity may be that the AYA cancers exhibit unique biologic characteristics, resulting in differences in clinical and treatment resistance behaviors. This report from the biologic component of the jointly sponsored National Cancer Institute and LiveStrong Foundation workshop entitled "Next Steps in Adolescent and Young Adult Oncology" summarizes the current status of biologic and translational research progress for 5 AYA cancers; colorectal cancer breast cancer, acute lymphoblastic leukemia, melanoma, and sarcoma. Conclusions from this meeting included the need for basic biologic, genomic, and model development for AYA cancers as well as translational research studies to elucidate any fundamental differences between pediatric, AYA, and adult cancers. The biologic questions for future research are whether there are mutational or signaling pathway differences (for example, between adult and AYA colorectal cancer) that can be clinically exploited to develop novel therapies for treating AYA cancers and to develop companion diagnostics.
Collapse
Affiliation(s)
- James V Tricoli
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland
| | - Donald G Blair
- Division of Cancer Biology, National Cancer Institute, Rockville, Maryland
| | - Carey K Anders
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - W Archie Bleyer
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, Oregon
| | - Lisa A Boardman
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Javed Khan
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Shivaani Kummar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland
| | - Brandon Hayes-Lattin
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, Oregon
| | - Stephen P Hunger
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Melinda Merchant
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Nita L Seibel
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland
| | - Magdalena Thurin
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland
| | | |
Collapse
|
25
|
Tao L, Gomez SL, Keegan THM, Kurian AW, Clarke CA. Breast Cancer Mortality in African-American and Non-Hispanic White Women by Molecular Subtype and Stage at Diagnosis: A Population-Based Study. Cancer Epidemiol Biomarkers Prev 2015; 24:1039-45. [PMID: 25969506 PMCID: PMC4490947 DOI: 10.1158/1055-9965.epi-15-0243] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/05/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Higher breast cancer mortality rates for African-American than non-Hispanic White women are well documented; however, it remains uncertain if this disparity occurs in disease subgroups defined by tumor molecular markers and stage at diagnosis. We examined racial differences in outcome according to subtype and stage in a diverse, population-based series of 103,498 patients. METHODS We obtained data for all invasive breast cancers diagnosed between January 1, 2005, and December 31, 2012, and followed through December 31, 2012, among 93,760 non-Hispanic White and 9,738 African-American women in California. Molecular subtypes were categorized according to tumor expression of hormone receptor (HR, based on estrogen and progesterone receptors) and human epidermal growth factor receptor 2 (HER2). Cox proportional hazards models were used to calculate relative hazard (RH) and 95% confidence intervals (CI) for breast cancer-specific mortality. RESULTS After adjustment for patient, tumor, and treatment characteristics, outcomes were comparable by race for stage I or IV cancer regardless of subtype, and HR(+)/HER2(+) or HR(-)/HER2(+) cancer regardless of stage. We found substantially higher hazards of breast cancer death among African-American women with stage II/III HR(+)/HER2(-) (RH, 1.31; 95% CI, 1.03-1.65; and RH, 1.39; 95% CI, 1.10-1.75, respectively) and stage III triple-negative cancers relative to Whites. CONCLUSIONS There are substantial racial/ethnic disparities among patients with stages II/III HR(+)/HER2(-) and stage III triple-negative breast cancers but not for other subtype and stage. IMPACT These data provide insights to assess barriers to targeted treatment (e.g., trastuzumab or endocrine therapy) of particular subtypes of breast cancer among African-American patients.
Collapse
Affiliation(s)
- Li Tao
- Cancer Prevention Institute of California, Fremont, California
| | - Scarlett Lin Gomez
- Cancer Prevention Institute of California, Fremont, California. Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California
| | - Theresa H M Keegan
- Cancer Prevention Institute of California, Fremont, California. Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California
| | - Allison W Kurian
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California. Medicine, Stanford University School of Medicine, Stanford, California
| | - Christina A Clarke
- Cancer Prevention Institute of California, Fremont, California. Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
26
|
Willis S, De P, Dey N, Long B, Young B, Sparano JA, Wang V, Davidson NE, Leyland-Jones BR. Enriched transcription factor signatures in triple negative breast cancer indicates possible targeted therapies with existing drugs. Meta Gene 2015; 4:129-41. [PMID: 26005638 PMCID: PMC4436509 DOI: 10.1016/j.mgene.2015.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 04/09/2015] [Accepted: 04/15/2015] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Triple negative (TN) breast cancers which lack expression of the estrogen (ER), progesterone (PR), and human epidermal growth factor 2 (HER2) receptors convey a poor prognosis due in part to a lack of targeted therapies. METHODS To identify viable targets for the treatment of TN disease, we have conducted a gene set enrichment analysis (GSEA) on seven different breast cancer whole genome gene expression cohorts comparing TN vs. ER + HER2 - to identify consistently enriched genes that share a common promoter motif. The seven cohorts were profiled on three different genome expression platforms (Affymetrix, Illumina and RNAseq) consisting in total of 2088 samples with IHC metadata. RESULTS GSEA identified enriched gene expression patterns in TN samples that share common promoter motifs associated with SOX9, E2F1, HIF1A, HMGA1, MYC BACH2, CEBPB, and GCNF/NR6A1. Unexpectedly, NR6A1 an orphan nuclear receptor normally expressed in germ cells of gonads is highly expressed in TN and ER + HER2 - samples making it an ideal drug target. CONCLUSION With the increasing number of large sample size breast cancer cohorts, an exploratory analysis of genes that are consistently enriched in TN sharing common promoter motifs allows for the identification of possible therapeutic targets with extensive validation in patient derived data sets.
Collapse
Affiliation(s)
| | - Pradip De
- Avera Cancer Institute, Sioux Falls, SD, United States
| | - Nandini Dey
- Avera Cancer Institute, Sioux Falls, SD, United States
| | - Bradley Long
- The Scripps Research Institute, Jupiter, FL, United States
| | - Brandon Young
- Avera Cancer Institute, Sioux Falls, SD, United States
| | | | - Victoria Wang
- Dana Farber Cancer Institute, Boston, MA, United States
| | - Nancy E Davidson
- University of Pittsburgh Cancer Institute and UPMC Cancer Center, Pittsburgh, PA, United States
| | | |
Collapse
|
27
|
Sturtz LA, Melley J, Mamula K, Shriver CD, Ellsworth RE. Outcome disparities in African American women with triple negative breast cancer: a comparison of epidemiological and molecular factors between African American and Caucasian women with triple negative breast cancer. BMC Cancer 2014; 14:62. [PMID: 24495414 PMCID: PMC3916697 DOI: 10.1186/1471-2407-14-62] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 02/02/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Although diagnosed less often, breast cancer in African American women (AAW) displays different characteristics compared to breast cancer in Caucasian women (CW), including earlier onset, less favorable clinical outcome, and an aggressive tumor phenotype. These disparities may be attributed to differences in socioeconomic factors such as access to health care, lifestyle, including increased frequency of obesity in AAW, and tumor biology, especially the higher frequency of triple negative breast cancer (TNBC) in young AAW. Improved understanding of the etiology and molecular characteristics of TNBC in AAW is critical to determining whether and how TNBC contributes to survival disparities in AAW. METHODS Demographic, pathological and survival data from AAW (n = 62) and CW (n = 98) with TNBC were analyzed using chi-square analysis, Student's t-tests, and log-rank tests. Frozen tumor specimens were available from 57 of the TNBC patients (n = 23 AAW; n = 34 CW); RNA was isolated after laser microdissection of tumor cells and was hybridized to HG U133A 2.0 microarrays. Data were analyzed using ANOVA with FDR <0.05, >2-fold difference defining significance. RESULTS The frequency of TNBC compared to all BC was significantly higher in AAW (28%) compared to CW (12%), however, significant survival and pathological differences were not detected between populations. Gene expression analysis revealed the tumors were more similar than different at the molecular level, with only CRYBB2P1, a pseudogene, differentially expressed between populations. Among demographic characteristics, AAW consumed significantly lower amounts of caffeine and alcohol, were less likely to breastfeed and more likely to be obese. CONCLUSIONS These data suggest that TNBC in AAW is not a unique disease compared to TNBC in CW. Rather, higher frequency of TNBC in AAW may, in part, be attributable to the effects of lifestyle choices. Because these risk factors are modifiable, they provide new opportunities for the development of risk reduction strategies that may decrease mortality by preventing the development of TNBC in AAW.
Collapse
Affiliation(s)
| | | | | | | | - Rachel E Ellsworth
- Clinical Breast Care Project, Henry M, Jackson Foundation for the Advancement of Military Medicine, Windber, PA, USA.
| |
Collapse
|
28
|
Chavez-MacGregor M, Liu S, De Melo-Gagliato D, Chen H, Do KA, Pusztai L, Symmans WF, Nair L, Hortobagyi GN, Mills GB, Meric-Bernstam F, Gonzalez-Angulo AM. Differences in gene and protein expression and the effects of race/ethnicity on breast cancer subtypes. Cancer Epidemiol Biomarkers Prev 2014; 23:316-23. [PMID: 24296856 PMCID: PMC3946290 DOI: 10.1158/1055-9965.epi-13-0929] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Differences in gene or protein expression patterns between breast cancers according to race/ethnicity and cancer subtype. METHODS Transcriptional profiling was performed using Affymetrix HG-U133A platform in 376 patients and reverse phase protein array analysis (RPPA) was done for 177 proteins in 255 patients from a separate cohort. Unsupervised clustering was conducted, as well as supervised comparison by race and tumor subtype. Standard statistical methods, BRB-Array tools, and Ingenuity Pathways software packages were used to analyze the data. RESULTS Median age was 50 years in both the cohorts. In the RPPA cohort, 54.5% of the tumors were hormone receptor-positive (HR-positive), 20.7% HER2-positive, and 24.71% triple-negative (TNBC). One hundred and forty-seven (57.6%), 47 (18.43%), and 46 (18.1%) of the patients were White, Hispanic, and Black, respectively. Unsupervised hierarchical clustering of the protein expression data showed no distinct clusters by race (P values were 0.492, 0.489, and 0.494 for the HR-positive, HER2-positive, and TNBC tumors respectively). In the gene expression cohort, 54.2% of the tumors were HR-positive, 16.5% HER2-positive, and 29.3% TNBC. Two hundred and sixteen (57.5%), 111 (29.52%), and 32 (8.52%) patients were White, Hispanic, and Black, respectively. No probe set with a false discovery rate (FDR) of <0.05 showed an association with race by breast cancer subtype; similar results were obtained using pathway and gene set enrichment analysis methods. CONCLUSIONS We did not detect a significant variation in RNA or protein expression comparing different race/ethnicity groups of women with breast cancer. IMPACT More research on the complex network of factors that result in outcomes differences among race/ethnicities is needed.
Collapse
Affiliation(s)
- Mariana Chavez-MacGregor
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shuying Liu
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Debora De Melo-Gagliato
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Huiqin Chen
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lajos Pusztai
- Division of Hematology-Oncology, Yale University, New Haven, CT, USA
| | - W. Fraser Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lakshmy Nair
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel N. Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gordon B. Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ana M. Gonzalez-Angulo
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
29
|
Lindner R, Sullivan C, Offor O, Lezon-Geyda K, Halligan K, Fischbach N, Shah M, Bossuyt V, Schulz V, Tuck DP, Harris LN. Molecular phenotypes in triple negative breast cancer from African American patients suggest targets for therapy. PLoS One 2013; 8:e71915. [PMID: 24260093 PMCID: PMC3832509 DOI: 10.1371/journal.pone.0071915] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 07/04/2013] [Indexed: 12/20/2022] Open
Abstract
Triple negative breast cancer (TNBC) is characterized by high proliferation, poor differentiation and a poor prognosis due to high rates of recurrence. Despite lower overall incidence African American (AA) patients suffer from higher breast cancer mortality in part due to the higher proportion of TNBC cases among AA patients compared to European Americans (EA). It was recently shown that the clinical heterogeneity of TNBC is reflected by distinct transcriptional programs with distinct drug response profiles in preclinical models. In this study, gene expression profiling and immunohistochemistry were used to elucidate potential differences between TNBC tumors of EA and AA patients on a molecular level. In a retrospective cohort of 136 TNBC patients, a major transcriptional signature of proliferation was found to be significantly upregulated in samples of AA ethnicity. Furthermore, transcriptional profiles of AA tumors showed differential activation of insulin-like growth factor 1 (IGF1) and a signature of BRCA1 deficiency in this cohort. Using signatures derived from the meta-analysis of TNBC gene expression carried out by Lehmann et al., tumors from AA patients were more likely of basal-like subtypes whereas transcriptional features of many EA samples corresponded to mesenchymal-like or luminal androgen receptor driven subtypes. These results were validated in The Cancer Genome Atlas mRNA and protein expression data, again showing enrichment of a basal-like phenotype in AA tumors and mesenchymal subtypes in EA tumors. In addition, increased expression of VEGF-activated genes together with elevated microvessel area determined by the AQUA method suggest that AA patients exhibit higher tumor vascularization. This study confirms the existence of distinct transcriptional programs in triple negative breast cancer in two separate cohorts and that these programs differ by racial group. Differences in TNBC subtypes and levels of tumor angiogenesis in AA versus EA patients suggest that targeted therapy choices should be considered in the context of race.
Collapse
MESH Headings
- Adult
- Black or African American
- Aged
- Breast Neoplasms/ethnology
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Databases, Genetic
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Middle Aged
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neovascularization, Pathologic/ethnology
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Neoplasm/biosynthesis
- RNA, Neoplasm/genetics
- Retrospective Studies
Collapse
Affiliation(s)
- Robert Lindner
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany
| | - Catherine Sullivan
- Medical Oncology, Yale Cancer Center, New Haven, Connecticut, United States of America
| | - Onyinye Offor
- Medical Oncology, Yale Cancer Center, New Haven, Connecticut, United States of America
| | - Kimberly Lezon-Geyda
- Medical Oncology, Yale Cancer Center, New Haven, Connecticut, United States of America
| | - Kyle Halligan
- Medical Oncology, Yale Cancer Center, New Haven, Connecticut, United States of America
| | - Neal Fischbach
- Department of Oncology, Bridgeport Hospital, Bridgeport, Connecticut, United States of America
| | - Mansi Shah
- Department of Oncology, Bridgeport Hospital, Bridgeport, Connecticut, United States of America
| | - Veerle Bossuyt
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Vincent Schulz
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - David P. Tuck
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Lyndsay N. Harris
- Medical Oncology, Yale Cancer Center, New Haven, Connecticut, United States of America
- University Hospitals, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
30
|
Danforth DN. Disparities in breast cancer outcomes between Caucasian and African American women: a model for describing the relationship of biological and nonbiological factors. Breast Cancer Res 2013; 15:208. [PMID: 23826992 PMCID: PMC3706895 DOI: 10.1186/bcr3429] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Breast cancer is the most common malignancy in women in the United States but significant disparities exist for African American women compared to Caucasian women. African American women present with breast cancer at a younger age and with a greater incidence under the age of 50 years, develop histologically more aggressive tumors that are at a more advanced stage at presentation, and have a worse disease-free and overall survival than Caucasian women. The biological characteristics of the primary tumor play an important role in determining the outcome of the disparity, and significant differences have been identified between African American and Caucasian breast cancer in steroid receptor and growth factor receptor content, mutations in cell cycle components, chromosomal abnormalities, and tumor suppressor and other cancer genes. The consequences of the biological factors are influenced by a variety of nonbiological factors, including socioeconomic, health care access, reproductive, and confounding factors. The nonbiological factors may act directly to enhance (or inhibit) the consequences of the biological changes, indirectly to facilitate outcome of the disparity, or as a cofounding factor, driving the association between the biological factors and the disparity. The prevention and management of the disparities will require an understanding of the relationship of biological and nonbiological factors. The present review was undertaken to promote this understanding by describing the biological basis of the four major disparities - early age of onset, more advanced stage of disease, more aggressive histologic changes, and worse survival - and the important relationship to the nonbiological factors. A model is proposed to provide a comprehensive view of this relationship, with the goal of facilitating an understanding of each disparity and the issues that need to be addressed to eliminate the disparity.
Collapse
Affiliation(s)
- David N Danforth
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
31
|
Ly M, Valent A, Diallo G, Penault-Lorca F, Dumke K, Marty V, Viehl P, Lazar V, Job B, Richon C, Scott V, Diallo DA, Bernaudin JF, Andre F. Gene copy number variations in breast cancer of Sub-Saharan African women. Breast 2013; 22:295-300. [PMID: 22999459 DOI: 10.1016/j.breast.2012.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 02/28/2012] [Accepted: 07/08/2012] [Indexed: 12/23/2022] Open
Abstract
The goal of this study was CGH array profiling of breast cancer from Malian women in order to define differences with those from USA. CGH array was performed in 28 samples, 17 with a triple negative phenotype. The profiles were compared to those of 106 tumors from USA. 6 chromosomal regions (6p21, 9q34, 11q13, 12q24, 17q25 and 22q12.1-22q13.1) were identified with a significant higher rate of copy number alterations. These regions contain several genes of interest including BCR. FISH and IHC confirmed that BCR was amplified and overexpressed particularly in triple negative tumors. Finally, 5 regions presented a high level of amplification in two or more samples, including 2 regions located between 9p22.3-9p23 and 9p23-9p24.1. This study confirms that breast cancers from African women present biological differences with those from USA. Larger studies are needed to go further in the identification of therapeutic targets that would be specific to African women.
Collapse
Affiliation(s)
- Madani Ly
- Service d'Hématologie et d'Oncologie Médicale, Hôpital du point G, Bamako, Mali
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Prognostic value of bcl-2 expression among women with breast cancer in Libya. Tumour Biol 2013; 34:1569-78. [PMID: 23417836 DOI: 10.1007/s13277-013-0687-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 02/03/2013] [Indexed: 12/21/2022] Open
Abstract
We studied the association of the immunohistochemical bcl-2 expression in Libyan breast cancer with clinicopathological variables and patient outcome. Histological samples from 170 previously untreated primary Libyan breast carcinoma patients were examined. In immunohistochemistry, the NCL-L-bcl-2-486 monoclonal antibody was used. Positive expression of bcl-2 was found in 106 patients (62.4 %). The bcl-2 expression was significantly associated with estrogen receptor (p<0.0001) and progesterone receptor positive tumors (p=0.002), small tumor size (p<0.0001), low tumor grade (p<0.0001), negative axillary lymph nodes (p<0.0001), early stages (p=0.001), and low risk of metastasis (p<0.0001). Positive expression was also associated with older patients (>50 years; p=0.04). Histological subtypes and family history of breast cancer did not have significant relationship with bcl-2. Patients with positive expression of bcl-2 had lower recurrence rate than bcl-2-negative patients and better survival after median follow-up of 47 months. Patients with high bcl-2 staining were associated with the best survival. The role of bcl-2 as an independent predictor of disease-specific survival was assessed in a multivariate survival (Cox) analysis, including age, hormonal status, recurrence, histological grade, and clinical stage variables. Bcl-2 (p<0.0001) and clinical stage (p=0.016) were independent predicators of disease-specific survival. For analysis of disease-free survival, the same variables were entered to the model and only bcl-2 proved to be an independent predictor (p=0.002). Patients with positive expression of bcl-2 were associated with low grade of malignancy, with lower recurrence rate, with lower rate of death, and with longer survival time. Bcl-2 is an independent predictor of breast cancer outcome, and it provides useful prognostic information in Libyan breast cancer. Thus, it could be used with classical clinicopathological factors to improve patient selection for therapy.
Collapse
|
33
|
Comparative analysis of methods for identifying recurrent copy number alterations in cancer. PLoS One 2012; 7:e52516. [PMID: 23285074 PMCID: PMC3527554 DOI: 10.1371/journal.pone.0052516] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 11/14/2012] [Indexed: 11/19/2022] Open
Abstract
Recurrent copy number alterations (CNAs) play an important role in cancer genesis. While a number of computational methods have been proposed for identifying such CNAs, their relative merits remain largely unknown in practice since very few efforts have been focused on comparative analysis of the methods. To facilitate studies of recurrent CNA identification in cancer genome, it is imperative to conduct a comprehensive comparison of performance and limitations among existing methods. In this paper, six representative methods proposed in the latest six years are compared. These include one-stage and two-stage approaches, working with raw intensity ratio data and discretized data respectively. They are based on various techniques such as kernel regression, correlation matrix diagonal segmentation, semi-parametric permutation and cyclic permutation schemes. We explore multiple criteria including type I error rate, detection power, Receiver Operating Characteristics (ROC) curve and the area under curve (AUC), and computational complexity, to evaluate performance of the methods under multiple simulation scenarios. We also characterize their abilities on applications to two real datasets obtained from cancers with lung adenocarcinoma and glioblastoma. This comparison study reveals general characteristics of the existing methods for identifying recurrent CNAs, and further provides new insights into their strengths and weaknesses. It is believed helpful to accelerate the development of novel and improved methods.
Collapse
|
34
|
Craig DW, O'Shaughnessy JA, Kiefer JA, Aldrich J, Sinari S, Moses TM, Wong S, Dinh J, Christoforides A, Blum JL, Aitelli CL, Osborne CR, Izatt T, Kurdoglu A, Baker A, Koeman J, Barbacioru C, Sakarya O, De La Vega FM, Siddiqui A, Hoang L, Billings PR, Salhia B, Tolcher AW, Trent JM, Mousses S, Von Hoff D, Carpten JD. Genome and transcriptome sequencing in prospective metastatic triple-negative breast cancer uncovers therapeutic vulnerabilities. Mol Cancer Ther 2012; 12:104-16. [PMID: 23171949 DOI: 10.1158/1535-7163.mct-12-0781] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) is characterized by the absence of expression of estrogen receptor, progesterone receptor, and HER-2. Thirty percent of patients recur after first-line treatment, and metastatic TNBC (mTNBC) has a poor prognosis with median survival of one year. Here, we present initial analyses of whole genome and transcriptome sequencing data from 14 prospective mTNBC. We have cataloged the collection of somatic genomic alterations in these advanced tumors, particularly those that may inform targeted therapies. Genes mutated in multiple tumors included TP53, LRP1B, HERC1, CDH5, RB1, and NF1. Notable genes involved in focal structural events were CTNNA1, PTEN, FBXW7, BRCA2, WT1, FGFR1, KRAS, HRAS, ARAF, BRAF, and PGCP. Homozygous deletion of CTNNA1 was detected in 2 of 6 African Americans. RNA sequencing revealed consistent overexpression of the FOXM1 gene when tumor gene expression was compared with nonmalignant breast samples. Using an outlier analysis of gene expression comparing one cancer with all the others, we detected expression patterns unique to each patient's tumor. Integrative DNA/RNA analysis provided evidence for deregulation of mutated genes, including the monoallelic expression of TP53 mutations. Finally, molecular alterations in several cancers supported targeted therapeutic intervention on clinical trials with known inhibitors, particularly for alterations in the RAS/RAF/MEK/ERK and PI3K/AKT/mTOR pathways. In conclusion, whole genome and transcriptome profiling of mTNBC have provided insights into somatic events occurring in this difficult to treat cancer. These genomic data have guided patients to investigational treatment trials and provide hypotheses for future trials in this irremediable cancer.
Collapse
Affiliation(s)
- David W Craig
- Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Tina E, Lindqvist BM, Gabrielson M, Lubovac Z, Wegman P, Wingren S. The mitochondrial transporter SLC25A43 is frequently deleted and may influence cell proliferation in HER2-positive breast tumors. BMC Cancer 2012; 12:350. [PMID: 22883974 PMCID: PMC3462703 DOI: 10.1186/1471-2407-12-350] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 07/31/2012] [Indexed: 01/08/2023] Open
Abstract
Background Overexpression of the human epidermal growth factor receptor (HER) 2 is associated with poor prognosis and shortened survival in breast cancer patients. HER2 is a potent activator of several signaling pathways that support cell survival, proliferation and metabolism. In HER2-positive breast cancer there are most likely unexplored proteins that act directly or indirectly downstream of well established pathways and take part in tumor development and treatment response. Methods In order to identify novel copy number variations (CNVs) in HER2-positive breast cancer whole-genome single nucleotide polymorphism (SNP) arrays were used. A PCR-based loss of heterozygosis (LOH) assay was conducted to verify presence of deletion in HER2-positive breast cancer cases but also in HER2 negative breast cancers, cervical cancers and lung cancers. Screening for mutations was performed using single-strand conformation polymorphism (SSCP) followed by PCR sequencing. Protein expression was evaluated with immunohistochemistry (IHC). Results A common deletion at chromosome Xq24 was found in 80% of the cases. This locus harbors the gene solute carrier (SLC) family 25A member 43 (SLC25A43) encoding for a mitochondrial transport protein. The LOH assay revealed presence of SLC25A43 deletion in HER2-positive (48%), HER2-negative (9%), cervical (42%) and lung (67%) cancers. HER2-positive tumors with negative or low SLC25A43 protein expression had significantly lower S-phase fraction compared to tumors with medium or high expression (P = 0.024). Conclusions We have found deletion in the SLC25A43 gene to be a common event in HER2-positive breast cancer as well as in other cancers. In addition, the SLC25A43 protein expression was shown to be related to S-phase fraction in HER2-positive breast cancer. Our results indicate a possible role of SLC25A43 in HER2-positive breast cancer and support the hypothesis of altered mitochondrial function in cancer.
Collapse
Affiliation(s)
- Elisabet Tina
- Clinical Research Centre, Örebro University Hospital, SE-70185, Örebro, Sweden.
| | | | | | | | | | | |
Collapse
|
36
|
Relationship between tumor DNA methylation status and patient characteristics in African-American and European-American women with breast cancer. PLoS One 2012; 7:e37928. [PMID: 22701537 PMCID: PMC3365111 DOI: 10.1371/journal.pone.0037928] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 04/30/2012] [Indexed: 01/06/2023] Open
Abstract
Aberrant DNA methylation is critical for development and progression of breast cancer. We investigated the association of CpG island methylation in candidate genes and clinicopathological features in 65 African-American (AA) and European-American (EA) breast cancer patients. Quantitative methylation analysis was carried out on bisulfite modified genomic DNA and sequencing (pyrosequencing) for promoter CpG islands of p16, ESR1, RASSF1A, RARβ2, CDH13, HIN1, SFRP1 genes and the LINE1 repetitive element using matched paired non-cancerous and breast tumor specimen (32 AA and 33 EA women). Five of the genes, all known tumor suppressor genes (RASSF1A, RARβ2, CDH13, HIN1 and SFRP1), were found to be frequently hypermethylated in breast tumor tissues but not in the adjacent non-cancerous tissues. Significant differences in the CDH13 methylation status were observed by comparing DNA methylation between AA and EA patients, with more obvious CDH13 methylation differences between the two patient groups in the ER- disease and among young patients (age<50). In addition, we observed associations between CDH13, SFRP1, and RASSF1A methylation and breast cancer subtypes and between SFRP1 methylation and patient's age. Furthermore, tumors that received neoadjuvant therapy tended to have reduced RASSF1A methylation when compared with chemotherapy naïve tumors. Finally, Kaplan Meier survival analysis showed a significant association between methylation at 3 loci (RASSF1A, RARβ2 and CDH13) and reduced overall disease survival. In conclusion, the DNA methylation status of breast tumors was found to be significantly associated with clinicopathological features and race/ethnicity of the patients.
Collapse
|