1
|
Demetriou C, Abid N, Butterworth M, Lezina L, Sandhu P, Howells L, Powley IR, Pringle JH, Sidat Z, Qassid O, Purnell D, Kaushik M, Duckworth K, Hartshorn H, Thomas A, Shaw JA, MacFarlane M, Pritchard C, Miles GJ. An optimised patient-derived explant platform for breast cancer reflects clinical responses to chemotherapy and antibody-directed therapy. Sci Rep 2024; 14:12833. [PMID: 38834809 DOI: 10.1038/s41598-024-63170-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 05/27/2024] [Indexed: 06/06/2024] Open
Abstract
Breast Cancer is the most common cancer among women globally. Despite significant improvements in overall survival, many tumours are refractory to therapy and so novel approaches are required to improve patient outcomes. We have evaluated patient-derived explants (PDEs) as a novel preclinical platform for breast cancer (BC) and implemented cutting-edge digital pathology and multi-immunofluorescent approaches for investigating biomarker changes in both tumour and stromal areas at endpoint. Short-term culture of intact fragments of BCs as PDEs retained an intact immune microenvironment, and tumour architecture was augmented by the inclusion of autologous serum in the culture media. Cell death/proliferation responses to FET chemotherapy in BC-PDEs correlated significantly with BC patient progression-free survival (p = 0.012 and p = 0.0041, respectively) and cell death responses to the HER2 antibody therapy trastuzumab correlated significantly with HER2 status (p = 0.018). These studies show that the PDE platform combined with digital pathology is a robust preclinical approach for informing clinical responses to chemotherapy and antibody-directed therapies in breast cancer. Furthermore, since BC-PDEs retain an intact tumour architecture over the short-term, they facilitate the preclinical testing of anti-cancer agents targeting the tumour microenvironment.
Collapse
Affiliation(s)
- Constantinos Demetriou
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, LE2 7LX, UK
| | - Naila Abid
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, LE2 7LX, UK
| | - Michael Butterworth
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, LE2 7LX, UK
| | - Larissa Lezina
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, LE2 7LX, UK
| | - Pavandeep Sandhu
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, LE2 7LX, UK
| | - Lynne Howells
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, LE2 7LX, UK
| | - Ian R Powley
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, LE2 7LX, UK
| | - James H Pringle
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, LE2 7LX, UK
| | - Zahirah Sidat
- HOPE Clinical Trials Facility, University Hospitals of Leicester NHS Trust, Sandringham Building, Leicester Royal Infirmary, Leicester, LE1 5WW, UK
| | - Omar Qassid
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, LE2 7LX, UK
- Pathology Department, University Hospitals of Leicester NHS Trust, Leicester Glenfield General Hospital, Groby Road, Leicester, LE3 9QP, UK
| | - Dave Purnell
- Pathology Department, University Hospitals of Leicester NHS Trust, Leicester Glenfield General Hospital, Groby Road, Leicester, LE3 9QP, UK
| | - Monika Kaushik
- Breast Care Centre, University Hospitals of Leicester NHS Trust, Leicester Glenfield General Hospital, Groby Road, Leicester, LE3 9QP, UK
| | - Kaitlin Duckworth
- Breast Care Centre, University Hospitals of Leicester NHS Trust, Leicester Glenfield General Hospital, Groby Road, Leicester, LE3 9QP, UK
| | - Helen Hartshorn
- Breast Care Centre, University Hospitals of Leicester NHS Trust, Leicester Glenfield General Hospital, Groby Road, Leicester, LE3 9QP, UK
| | - Anne Thomas
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, LE2 7LX, UK
| | - Jacqui A Shaw
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, LE2 7LX, UK
| | - Marion MacFarlane
- MRC Toxicology Unit, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK.
- Department of Molecular and Cell Biology, University of Leicester, Leicester, LE1 7HB, UK.
| | - Catrin Pritchard
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, LE2 7LX, UK.
| | - Gareth J Miles
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, LE2 7LX, UK.
| |
Collapse
|
2
|
Lan HR, Chen M, Yao SY, Chen JX, Jin KT. Novel immunotherapies for breast cancer: Focus on 2023 findings. Int Immunopharmacol 2024; 128:111549. [PMID: 38266449 DOI: 10.1016/j.intimp.2024.111549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Immunotherapy has emerged as a revolutionary approach in cancer therapy, and recent advancements hold significant promise for breast cancer (BCa) management. Employing the patient's immune system to combat BCa has become a focal point in immunotherapeutic investigations. Strategies such as immune checkpoint inhibitors (ICIs), adoptive cell transfer (ACT), and targeting the tumor microenvironment (TME) have disclosed encouraging clinical outcomes. ICIs, particularly programmed cell death protein 1 (PD-1)/PD-L1 inhibitors, exhibit efficacy in specific BCa subtypes, including triple-negative BCa (TNBC) and human epidermal growth factor receptor 2 (HER2)-positive cancers. ACT approaches, including tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR) T-cell therapy, showed promising clinical outcomes in enhancing tumor recognition and elimination. Targeting the TME through immune agonists and oncolytic viruses signifies a burgeoning field of research. While challenges persist in patient selection, resistance mechanisms, and combination therapy optimization, these novel immunotherapies hold transformative potential for BCa treatment. Continued research and clinical trials are imperative to refine and implement these innovative approaches, paving the way for improved outcomes and revolutionizing the management of BCa. This review provides a concise overview of the latest immunotherapies (2023 studies) in BCa, highlighting their potential and current status.
Collapse
Affiliation(s)
- Huan-Rong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China
| | - Min Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Shi-Ya Yao
- Department of Gastrointestinal, Colorectal and Anal Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China
| | - Jun-Xia Chen
- Department of Gynecology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, China.
| | - Ke-Tao Jin
- Department of Gastrointestinal, Colorectal and Anal Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China.
| |
Collapse
|
3
|
Obeagu EI, Obeagu GU. Breast cancer: A review of risk factors and diagnosis. Medicine (Baltimore) 2024; 103:e36905. [PMID: 38241592 PMCID: PMC10798762 DOI: 10.1097/md.0000000000036905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 12/18/2023] [Indexed: 01/21/2024] Open
Abstract
Breast cancer remains a complex and prevalent health concern affecting millions of individuals worldwide. This review paper presents a comprehensive analysis of the multifaceted landscape of breast cancer, elucidating the diverse spectrum of risk factors contributing to its occurrence and exploring advancements in diagnostic methodologies. Through an extensive examination of current literature, various risk factors have been identified, encompassing genetic predispositions such as BRCA mutations, hormonal influences, lifestyle factors, and reproductive patterns. Age, family history, and environmental factors further contribute to the intricate tapestry of breast cancer etiology. Moreover, this review delineates the pivotal role of diagnostic tools in the early detection and management of breast cancer. Mammography, the cornerstone of breast cancer screening, is augmented by emerging technologies like magnetic resonance imaging and molecular testing, enabling improved sensitivity and specificity in diagnosing breast malignancies. Despite these advancements, challenges persist in ensuring widespread accessibility to screening programs, particularly in resource-limited settings. In conclusion, this review underscores the importance of understanding diverse risk factors in the development of breast cancer and emphasizes the critical role of evolving diagnostic modalities in enhancing early detection. The synthesis of current knowledge in this review aims to contribute to a deeper comprehension of breast cancer's multifactorial nature and inform future directions in research, screening strategies, and preventive interventions.
Collapse
|
4
|
Thanasan S, Sukhakul K, Chitpakdee S, Kitkumthorn N. Diagnostic Accuracy of Immunohistochemistry for HER2-Positive Breast Cancer. Asian Pac J Cancer Prev 2023; 24:4321-4327. [PMID: 38156869 PMCID: PMC10909106 DOI: 10.31557/apjcp.2023.24.12.4321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024] Open
Abstract
OBJECTIVE Currently, human epidermal growth factor receptor 2 (HER2)-positive breast cancer cells are diagnosed under the American Society of Clinical Oncology (ASCO) and College of American Pathologists (CAP) 2018 guidelines. The guideline combined the results of in situ hybridization (DISH) and immunohistochemistry (IHC) techniques. The IHC technique is easy, cheap, and suitable for developing country. Therefore, in this study, we validated the use of IHC alone compared to the results of HER2 amplification under ASCO/CAP 2018 guidelines in diagnosed HER2 positive breast cancer cells. METHODS A total of 510 breast cancer tissue samples from Rajavithi Hospital in Bangkok, Thailand, from January 1st, 2022, to May 31st, 2023, were analyzed by IHC, followed by dual ISH (DISH). We selected 58 samples of IHC equivocal (score 2+) and 98 samples of IHC positive (score 3+) to analyze the diagnostic values by comparing them to the results of HER2 amplification. RESULTS The HER2 IHC score was found to agree with HER2 amplification with a sensitivity of 87.96%, a specificity of 93.75%, a positive predictive value of 96.94%, a negative predictive value of 77.59%, a positive likelihood ratio of 14.07, a negative likelihood ratio of 0.13, and an accuracy of 89.74%. CONCLUSION The promising outcomes suggest that a positive IHC test result (score 3+) could potentially stand alone for patients with breast cancer undergoing anti-HER2 treatment, even without DISH confirmation.
Collapse
Affiliation(s)
- Siwaporn Thanasan
- Pathology Department, Rajavithi Hospital, Phayathai Road, Ratchathewi, Bangkok, Thailand.
| | - Kweekrong Sukhakul
- Pathology Department, Rajavithi Hospital, Phayathai Road, Ratchathewi, Bangkok, Thailand.
| | - Sakchai Chitpakdee
- Pathology Department, Rajavithi Hospital, Phayathai Road, Ratchathewi, Bangkok, Thailand.
| | - Nakarin Kitkumthorn
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
5
|
Vyas D, Wairkar S. Effect of variables on exemestane-loaded albumin nanoparticles: statistical optimization and anti-cancer activity in MCF-7 cell lines. Pharm Dev Technol 2023; 28:1048-1055. [PMID: 37987762 DOI: 10.1080/10837450.2023.2285925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023]
Abstract
This research aimed to evaluate the effect of variables on exemestane-loaded bovine serum albumin nanoparticles (EXE-BSA NPs) to improve anti-breast cancer activity. EXE-BSA NPs were optimized using 32 factorial design, wherein the concentration of BSA (X1) and sonication time (X2) were independent variables and particle size (Y1) and %w/w entrapment efficiency (Y2) were dependent variables. The statistical optimization revealed a significant effect of BSA concentration on both variables, whereas sonication time affected only particle size. The optimized EXE-BSA NPs were spherical with 124.1 ± 2.62 nm particle size, 83.95 ± 1.06% w/w drug entrapment, and exhibited a biphasic release of 100% (w/w) drug over 72 h. The optimized formulation induced cytotoxicity in MCF-7 cell lines with an IC50 value of 21.46 µg/mL by MTT assay, almost half the free drug (54.87 µg/mL). Thus, statistically optimized EXE-BSA NPs were effective in MCF-7 cell lines and can be explored to treat estrogen receptor-positive breast cancer.
Collapse
Affiliation(s)
- Darshan Vyas
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-University, Mumbai, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-University, Mumbai, India
| |
Collapse
|
6
|
Barrios-Rodríguez R, Garde C, Pérez-Carrascosa FM, Expósito J, Peinado FM, Fernández Rodríguez M, Requena P, Salcedo-Bellido I, Arrebola JP. Associations of accumulated persistent organic pollutants in breast adipose tissue with the evolution of breast cancer after surgery. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 897:165373. [PMID: 37419338 DOI: 10.1016/j.scitotenv.2023.165373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/09/2023]
Abstract
Chronic exposure to persistent organic pollutants (POPs) is suspected to contribute to the onset of breast cancer, but the impact on the evolution of patients after diagnosis is unclear. We aimed to analyze the contribution of long-term exposure to five POPs to overall mortality, cancer recurrence, metastasis, and development of second primary tumors over a global follow-up of 10 years after surgery in breast cancer patients in a cohort study. Between 2012 and 2014, a total of 112 newly diagnosed breast cancer patients were recruited from a public hospital in Granada, Southern Spain. Historical exposure to POPs was estimated by analyzing their concentrations in breast adipose tissue samples. Sociodemographic data were collected through face-to-face interviews, while data on evolution tumor were retrieved from clinical records. Statistical analyses were performed using Cox regression (overall survival, breast cancer recurrence or metastasis) and binary logistic regression models (joint outcome variable). We also tested for statistical interactions of POPs with age, residence, and prognostic markers. The third vs first tertile of hexachlorobenzene concentrations was associated with a lower risk of all-cause mortality (Hazard Ratio, HR = 0.26; 95 % Confidence Interval, CI = 0.07-0.92) and of the appearance of any of the four events (Odds Ratio = 0.37; 95 % CI = 0.14-1.03). Polychlorinated biphenyl 138 concentrations were significantly and inversely associated with risk of metastasis (HR = 0.65; 95 % CI = 0.44-0.97) and tumor recurrence (HR = 0.69; 95 % CI = 0.49-0.98). Additionally, p,p'-dichlorodiphenyldichloroethylene showed inverse associations with risk of metastasis in women with ER-positive tumors (HR = 0.49; 95 % CI = 0.25-0.93) and in those with a tumor size <2.0 cm (HR = 0.39; 95 % CI = 0.18-0.87). The observed paradoxical inverse associations of POP exposure with breast cancer evolution might be related to either a better prognosis of hormone-dependent tumors, which have an approachable pharmacological target, or an effect of sequestration of circulating POPs by adipose tissue.
Collapse
Affiliation(s)
- R Barrios-Rodríguez
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - C Garde
- San Cecilio University Hospital, Avenida del Conocimiento s/n, 18016 Granada, Spain
| | - F M Pérez-Carrascosa
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - J Expósito
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Virgen de las Nieves University Hospital, Radiation Oncology Department, Oncology Unit, Granada, Spain
| | - F M Peinado
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - M Fernández Rodríguez
- Universidad de Granada, Facultad de Farmacia, Departamento de Farmacia y Tecnología Farmacéutica, Granada, Spain
| | - P Requena
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - I Salcedo-Bellido
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain.
| | - J P Arrebola
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain.
| |
Collapse
|
7
|
Moyer CL, Brown PH. Targeting nuclear hormone receptors for the prevention of breast cancer. Front Med (Lausanne) 2023; 10:1200947. [PMID: 37583424 PMCID: PMC10424511 DOI: 10.3389/fmed.2023.1200947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/30/2023] [Indexed: 08/17/2023] Open
Abstract
Advancements in research have led to the steady decline of breast cancer mortality over the past thirty years. However, breast cancer incidence has continued to rise, resulting in an undue burden on healthcare costs and highlighting a great need for more effective breast cancer prevention strategies, including targeted chemo preventative agents. Efforts to understand the etiology of breast cancer have uncovered important roles for nuclear receptors in the development and progression of breast cancer. Targeted therapies to inhibit estrogen receptor (ER) and progesterone receptor (PR) signaling (selective ER modulators, aromatase inhibitors and selective PR modulators) have shown great promise for the treatment and prevention of hormone receptor (HR)-positive breast cancer. However, these drugs do not prevent HR-negative disease. Therefore, recent efforts have focused on novel targeted therapies with the potential to prevent both HR-positive and HR-negative breast cancer. Among these include drugs that target other nuclear receptors, such as retinoic acid receptor (RAR), retinoid X receptor (RXR) and vitamin D receptor (VDR). In this review we provide an overview of recent preclinical and clinical trials targeting members of the nuclear receptor superfamily for the prevention of breast cancer.
Collapse
Affiliation(s)
- Cassandra L. Moyer
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Powel H. Brown
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
8
|
González-Palacios Torres C, Barrios-Rodríguez R, Muñoz-Bravo C, Toledo E, Dierssen T, Jiménez-Moleón JJ. Mediterranean diet and risk of breast cancer: An umbrella review. Clin Nutr 2023; 42:600-608. [PMID: 36893621 DOI: 10.1016/j.clnu.2023.02.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 01/27/2023] [Accepted: 02/15/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND The Mediterranean Diet (MedDiet) is a healthy dietary pattern which has been related to a lower risk of certain chronic diseases, such as some cancers. However, its role in breast cancer development remains unclear. This umbrella review aims to summarize the highest available evidence on MedDiet and breast cancer risk. METHODS Pubmed, Web of Science, and Scopus electronic platforms were searched for relevant systematic reviews and meta-analyses. The selection criteria included systematic reviews with or without meta-analysis including women aged 18 years or older which evaluated the adherence to a MedDiet as the exposure and incidence of breast cancer as the outcome variable. Overlapping and quality of the reviews using AMSTAR-2 tool were independently assessed by two authors. RESULTS Five systematic reviews and six systematic reviews with meta-analysis were included. Overall, 4 systematic reviews - two with and two without meta-analysis - were rated as of high quality. An inverse association was found in 5 of the 9 reviews which evaluated the role of MedDiet on the risk of total breast cancer. The meta-analyses showed moderate-high heterogeneity. The risk reduction seemed to be more consistent among postmenopausal women. No association was found for MedDiet among premenopausal women. CONCLUSIONS The results of this umbrella review suggest that adherence to a MedDiet pattern had a protective effect on the risk of breast cancer, especially for postmenopausal breast cancer. The stratification of breast cancer cases and conducting high-quality reviews are aspects needed to overcome the current results' heterogeneity and to improve knowledge in this field.
Collapse
Affiliation(s)
| | - Rocío Barrios-Rodríguez
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; CIBER de Epidemiología y Salud Pública, Instituto de Salud Carlos III, Spain
| | - Carlos Muñoz-Bravo
- Department of Public Health and Psychiatry, School of Medicine, University of Málaga, Málaga, Spain; Biomedical Research Institute of Malaga (IBIMA), Málaga, Spain.
| | - Estefanía Toledo
- Department of Preventive Medicine and Public Health, University of Navarra-School of Medicine, Pamplona, Navarra, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Navarra, Spain
| | - Trinidad Dierssen
- CIBER de Epidemiología y Salud Pública, Instituto de Salud Carlos III, Spain; Universidad de Cantabria - IDIVAL, Santander, Spain
| | - José Juan Jiménez-Moleón
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; CIBER de Epidemiología y Salud Pública, Instituto de Salud Carlos III, Spain
| |
Collapse
|
9
|
Starek-Świechowicz B, Budziszewska B, Starek A. Alcohol and breast cancer. Pharmacol Rep 2023; 75:69-84. [PMID: 36310188 PMCID: PMC9889462 DOI: 10.1007/s43440-022-00426-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 02/04/2023]
Abstract
Breast cancer is one of the main causes of death in women worldwide. In women, breast cancer includes over half of all tumours caused by alcohol. This paper discusses both ethanol metabolism and the mechanisms of mammary tumourigenesis caused by alcohol. Numerous signalling pathways in neoplastic transformation following alcohol consumption in women have been presented. In addition, primary and secondary prevention, phytochemicals, synthetic chemicals, specific inhibitors of enzymes and selective receptor modulators have been described.
Collapse
Affiliation(s)
- Beata Starek-Świechowicz
- Department of Biochemical Toxicology, Chair of Toxicology, Medical College, Jagiellonian University, Medyczna 9, 30-688 Kraków, Poland
| | - Bogusława Budziszewska
- Department of Biochemical Toxicology, Chair of Toxicology, Medical College, Jagiellonian University, Medyczna 9, 30-688 Kraków, Poland
| | - Andrzej Starek
- Department of Biochemical Toxicology, Chair of Toxicology, Medical College, Jagiellonian University, Medyczna 9, 30-688 Kraków, Poland
| |
Collapse
|
10
|
Breast cancer prevention by short-term inhibition of TGFβ signaling. Nat Commun 2022; 13:7558. [PMID: 36476730 PMCID: PMC9729304 DOI: 10.1038/s41467-022-35043-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer prevention has a profound impact on cancer-associated mortality and morbidity. We previously identified TGFβ signaling as a candidate regulator of mammary epithelial cells associated with breast cancer risk. Here, we show that short-term TGFBR inhibitor (TGFBRi) treatment of peripubertal ACI inbred and Sprague Dawley outbred rats induces lasting changes and prevents estrogen- and carcinogen-induced mammary tumors, respectively. We identify TGFBRi-responsive cell populations by single cell RNA-sequencing, including a unique epithelial subpopulation designated secretory basal cells (SBCs) with progenitor features. We detect SBCs in normal human breast tissues and find them to be associated with breast cancer risk. Interactome analysis identifies SBCs as the most interactive cell population and the main source of insulin-IGF signaling. Accordingly, inhibition of TGFBR and IGF1R decrease proliferation of organoid cultures. Our results reveal a critical role for TGFβ in regulating mammary epithelial cells relevant to breast cancer and serve as a proof-of-principle cancer prevention strategy.
Collapse
|
11
|
Yousefi H, Bahramy A, Zafari N, Delavar MR, Nguyen K, Haghi A, Kandelouei T, Vittori C, Jazireian P, Maleki S, Imani D, Moshksar A, Bitaraf A, Babashah S. Notch signaling pathway: a comprehensive prognostic and gene expression profile analysis in breast cancer. BMC Cancer 2022; 22:1282. [PMID: 36476410 PMCID: PMC9730604 DOI: 10.1186/s12885-022-10383-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is a complex disease exhibiting a great degree of heterogeneity due to different molecular subtypes. Notch signaling regulates the differentiation of breast epithelial cells during normal development and plays a crucial role in breast cancer progression through the abnormal expression of the Notch up-and down-stream effectors. To date, there are only a few patient-centered clinical studies using datasets characterizing the role of Notch signaling pathway regulators in breast cancer; thus, we investigate the role and functionality of these factors in different subtypes using publicly available databases containing records from large studies. High-throughput genomic data and clinical information extracted from TCGA were analyzed. We performed Kaplan-Meier survival and differential gene expression analyses using the HALLMARK_NOTCH_SIGNALING gene set. To determine if epigenetic regulation of the Notch regulators contributes to their expression, we analyzed methylation levels of these factors using the TCGA HumanMethylation450 Array data. Notch receptors and ligands expression is generally associated with the tumor subtype, grade, and stage. Furthermore, we showed gene expression levels of most Notch factors were associated with DNA methylation rate. Modulating the expression levels of Notch receptors and effectors can be a potential therapeutic approach for breast cancer. As we outline herein, elucidating the novel prognostic and regulatory roles of Notch implicate this pathway as an essential mediator controlling breast cancer progression.
Collapse
Affiliation(s)
- Hassan Yousefi
- Biochemistry & Molecular Biology, Louisiana State University Health Science Center (LSUHSC), New Orleans, LA, USA
| | - Afshin Bahramy
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Narges Zafari
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Rostamian Delavar
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Khoa Nguyen
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Atousa Haghi
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Kandelouei
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Cecilia Vittori
- Louisiana State University Health Sciences Center (LSUHSC), and Stanley S. Scott Cancer Center, New Orleans, LA, USA
| | - Parham Jazireian
- Department of Biology, University Campus 2, University of Guilan, Rasht, Iran
| | - Sajad Maleki
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Danyal Imani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Amin Moshksar
- Interventional Radiology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Amirreza Bitaraf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box, Tehran, 14115-154, Iran
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box, Tehran, 14115-154, Iran.
| |
Collapse
|
12
|
Thabet RH, Gomaa AA, Matalqah LM, Shalaby EM. Vitamin D: an essential adjuvant therapeutic agent in breast cancer. J Int Med Res 2022; 50:3000605221113800. [PMID: 35883275 PMCID: PMC9340350 DOI: 10.1177/03000605221113800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 06/07/2022] [Indexed: 11/20/2022] Open
Abstract
Low serum levels of vitamin D have been reported as a risk factor for breast cancer. This narrative review provides an update on the impact of vitamin D on hormone receptors, notably estrogen receptor subunits, and gives insights on possible therapeutic interventions to overcome breast cancer. In addition, evidence that supports the beneficial use of vitamin D as adjuvant treatment of breast cancer is summarized. Vitamin D deficiency is significantly widespread in patients with triple-negative tumors. Several studies have observed a possible modulatory effect of vitamin D or its analogues on the expression of different hormone receptors in breast cancer and increased sensitivity to tamoxifen. Vitamin D possesses anti-inflammatory and immunomodulatory effects in patients with breast cancer, and the mechanism of action of vitamin D in patients with breast cancer is discussed. In conclusion, vitamin D appears to have a beneficial role in the prevention and management of breast cancer, however, large-scale, randomized controlled trials are needed to confirm the effects of vitamin D in breast cancer prevention or treatment.
Collapse
Affiliation(s)
- Romany H Thabet
- Department of Pharmacology, Faculty of Medicine, Assiut
University
- Department of Basic Medical Sciences, Faculty of Medicine,
Yarmouk University, Irbid-Jordan
| | - Adel A Gomaa
- Department of Pharmacology, Faculty of Medicine, Assiut
University
- Center for Research on Management of Age-Related Diseases,
Assiut University, Assiut, Egypt
| | - Laila M Matalqah
- Department of Basic Medical Sciences, Faculty of Medicine,
Yarmouk University, Irbid-Jordan
| | | |
Collapse
|
13
|
Saddiq AA, El-Far AH, Mohamed Abdullah SA, Godugu K, Almaghrabi OA, Mousa SA. Curcumin, thymoquinone, and 3, 3′-diindolylmethane combinations attenuate lung and liver cancers progression. Front Pharmacol 2022; 13:936996. [PMID: 35847018 PMCID: PMC9277483 DOI: 10.3389/fphar.2022.936996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/14/2022] [Indexed: 12/20/2022] Open
Abstract
Cancer can develop due to abnormal cell proliferation in any body’s cells, so there are over a hundred different types of cancer, each with its distinct behavior and response to treatment. Therefore, many studies have been conducted to slow cancer progression and find effective and safe therapies. Nutraceuticals have great attention for their anticancer potential. Therefore, the current study was conducted to investigate the anticancer effects of curcumin (Cur), thymoquinone (TQ), and 3, 3′-diindolylmethane (DIM) combinations on lung (A549) and liver (HepG2) cancer cell lines’ progression. Results showed that triple (Cur + TQ + DIM) and double (Cur + TQ, Cur + DIM, and TQ + DIM) combinations of Cur, TQ, and DIM significantly increased apoptosis with elevation of caspase-3 protein levels. Also, these combinations exhibited significantly decreased cell proliferation, migration, colony formation activities, phosphatidylinositol 3-kinase (PI3K), and protein kinase B (AKT) protein levels with S phase reduction. Triple and double combinations of Cur, TQ, and DIM hindered tumor weight and angiogenesis of A549 and HepG2 implants in the chorioallantoic membrane model. Interestingly, Cur, TQ, and DIM combinations are considered promising for suppressing cancer progression via inhibiting tumor angiogenesis. Further preclinical and clinical investigations are warranted.
Collapse
Affiliation(s)
- Amna A. Saddiq
- College of Sciences, Department of Biology, University of Jeddah, Jeddah, Saudi Arabia
| | - Ali H. El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
- *Correspondence: Ali H. El-Far,
| | - Shymaa Abdullah Mohamed Abdullah
- Molecular Biology Unit, Medical Technology Center and Applied Medical Chemistry Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Kavitha Godugu
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, United States
| | - Omar A. Almaghrabi
- College of Sciences, Department of Biology, University of Jeddah, Jeddah, Saudi Arabia
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, United States
| |
Collapse
|
14
|
Zhang Y, Yang X, Cui Y, Zhang X. Suppression of RNA editing by miR-17 inhibits the stemness of melanoma stem cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:439-455. [PMID: 35036056 PMCID: PMC8728536 DOI: 10.1016/j.omtn.2021.12.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022]
Abstract
More and more evidence suggests that microRNA (miRNA) and RNA editing play key roles in the development and progression of tumor. However, the influence of miRNA-mediated RNA editing on tumor stem cells remains unclear. In this study, the results demonstrated that miR-17, which was downregulated in melanoma stem cells, acted as a tumor inhibitor by suppressing the stemness of melanoma stem cells and promoting cell differentiation. MiR-17 targeted ADAR2 (adenosine deaminase acting on RNA 2), a gene encoding an editing enzyme required for the maintenance of melanoma stem cell stemness. In melanoma stem cells, ADAR2 was responsible for DOCK2 mRNA editing, which was able to increase the stability of DOCK2 mRNA. The in vitro and in vivo data demonstrated that DOCK2 mRNA editing upregulated the expressions of stemness and anti-apoptotic genes by activating Rac1 and then phosphorylating Akt and NF-κB, thus leading to oncogenesis of melanoma stem cells. Our findings contribute new perspectives to miRNA-regulated RNA editing in tumor progression.
Collapse
Affiliation(s)
- Yu Zhang
- College of Life Sciences, Laboratory for Marine Biology and Biotechnology of Pilot National Laboratory for Marine Science and Technology (Qingdao) and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhejiang University, Hangzhou 310058, People’s Republic of China
| | - Xiaoyuan Yang
- College of Life Sciences, Laboratory for Marine Biology and Biotechnology of Pilot National Laboratory for Marine Science and Technology (Qingdao) and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhejiang University, Hangzhou 310058, People’s Republic of China
| | - Yalei Cui
- College of Life Sciences, Laboratory for Marine Biology and Biotechnology of Pilot National Laboratory for Marine Science and Technology (Qingdao) and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhejiang University, Hangzhou 310058, People’s Republic of China
| | - Xiaobo Zhang
- College of Life Sciences, Laboratory for Marine Biology and Biotechnology of Pilot National Laboratory for Marine Science and Technology (Qingdao) and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhejiang University, Hangzhou 310058, People’s Republic of China
| |
Collapse
|
15
|
El-Far AH, Saddiq AA, Mohamed SA, Almaghrabi OA, Mousa SA. Curcumin and Thymoquinone Combination Attenuates Breast Cancer Cell Lines' Progression. Integr Cancer Ther 2022; 21:15347354221099537. [PMID: 35583244 PMCID: PMC9128062 DOI: 10.1177/15347354221099537] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Breast cancer is the most harmful malignancy in women worldwide. Therefore, in the current study, we investigated the combinatory effect of natural bioactive compounds, including curcumin (Cur) and thymoquinone (TQ), on MCF7 and MDA-MB-231 breast cancer cell lines’ progression. We investigated the Fa values and combination index of Cur and TQ in this context. Moreover, cytotoxicity percentages, annexin-V, proliferation, colony formation, and migration assays were used along with cell cycle analysis. In addition, caspase-3, phosphatidylinositol 3-kinase (PI3K), and protein kinase B (AKT) protein levels were determined by ELISA assessment. The results showed that Cur, TQ, and Cur + TQ induced apoptosis with cell cycle arrest and decreased cell proliferation, colony formation, and migration activities. Cur + TQ combination significantly increased caspase-3 and decreased PI3K and AKT protein levels. These results suggest the promising anticancer benefit of the Cur and TQ combination against breast cancer.
Collapse
Affiliation(s)
| | | | | | | | - Shaker A Mousa
- Albany College of Pharmacy & Health Sciences, Rensselaer, NY, USA
| |
Collapse
|
16
|
Manrique-Moreno M, Santa-González G, Gallego V. Bioactive cationic peptides as potential agents for breast cancer treatment. Biosci Rep 2021; 41:BSR20211218C. [PMID: 34874400 PMCID: PMC8655503 DOI: 10.1042/bsr20211218c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/25/2021] [Accepted: 11/10/2021] [Indexed: 11/17/2022] Open
Abstract
Breast cancer continues to affect millions of women worldwide, and the number of new cases dramatically increases every year. The physiological causes behind the disease are still not fully understood. One in every 100 cases can occur in men, and although the frequency is lower than among women, men tend to have a worse prognosis of the disease. Various therapeutic alternatives to combat the disease are available. These depend on the type and progress of the disease, and include chemotherapy, radiotherapy, surgery, and cancer immunotherapy. However, there are several well-reported side effects of these treatments that have a significant impact on life quality, and patients either relapse or are refractory to treatment. This makes it necessary to develop new therapeutic strategies. One promising initiative are bioactive peptides, which have emerged in recent years as a family of compounds with an enormous number of clinical applications due to their broad spectrum of activity. They are widely distributed in several organisms as part of their immune system. The antitumoral activity of these peptides lies in a nonspecific mechanism of action associated with their interaction with cancer cell membranes, inducing, through several routes, bilayer destabilization and cell death. This review provides an overview of the literature on the evaluation of cationic peptides as potential agents against breast cancer under different study phases. First, physicochemical characteristics such as the primary structure and charge are presented. Secondly, information about dosage, the experimental model used, and the mechanism of action proposed for the peptides are discussed.
Collapse
Affiliation(s)
- Marcela Manrique-Moreno
- Chemistry Institute, Faculty of Exact and Natural Sciences, University of Antioquia, A.A. 1226, Medellin, Antioquia
| | - Gloria A. Santa-González
- Biomedical Innovation and Research Group, Faculty of Applied and Exact Sciences, Instituto Tecnólogico Metropolitano, A.A. 54959, Medellin, Colombia
| | - Vanessa Gallego
- Chemistry Institute, Faculty of Exact and Natural Sciences, University of Antioquia, A.A. 1226, Medellin, Antioquia
| |
Collapse
|
17
|
DHA inhibits Gremlin-1-induced epithelial-to-mesenchymal transition via ERK suppression in human breast cancer cells. Biosci Rep 2021; 40:222308. [PMID: 32141512 PMCID: PMC7087330 DOI: 10.1042/bsr20200164] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/22/2022] Open
Abstract
Docosahexaenoic acid (DHA) is an omega-3 fatty acid abundant in fish oils. It is known to have an inhibitory effect on various diseases such as inflammation, diabetes, and cancer. Epithelial-to-mesenchymal transition (EMT) is a process that epithelial cells gain migratory property to become mesenchymal cells involved in wound healing, organ fibrosis, and cancer progression. Gremlin-1 (GREM1) is a bone morphogenetic protein antagonist known to play a role in EMT. However, the role of GREM1 in the induction of EMT in human breast cancer cells and the effect of DHA on GREM1-induced EMT remain unclear. Establishment of GREM1 knockdown cell lines was performed using lentiviral shRNAs. Expression of EMT markers was determined by qRT-PCR and Western blotting. Effect of GREM1 and/or DHA on cell migration was investigated using wound healing assay. The level of GREM1 expression in human breast cancer tissues was determined by Oncomine database mining. GREM1 induced the expression of genes including N-cadherin, vimentin, and Slug. GREM1 promoted the migration of human breast cancer cells. GREM1 enhanced the expression of phosphorylated extracellular signal-regulated kinase (p-ERK) and the ERK activation was involved in EMT. Interestingly, DHA reduced the expression of GREM1. DHA also inhibited the expression of mesenchymal cell-associated genes and cell migration induced by GREM1. Furthermore, DHA suppressed the expression of p-ERK induced by GREM1. These results indicate that GREM1–ERK axis plays a role in EMT in human breast cancer cells and DHA is a putative compound that can inhibit EMT by inhibiting GREM1 signal transduction.
Collapse
|
18
|
Lee HS, Lee IH, Kang K, Park SI, Moon SJ, Lee CH, Lee DY. A Network Pharmacology Study on the Molecular Mechanisms of FDY003 for Breast Cancer Treatment. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:3919143. [PMID: 33628298 PMCID: PMC7881938 DOI: 10.1155/2021/3919143] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 01/25/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
Herbal medicines have drawn considerable attention with regard to their potential applications in breast cancer (BC) treatment, a frequently diagnosed malignant disease, considering their anticancer efficacy with relatively less adverse effects. However, their mechanisms of systemic action have not been understood comprehensively. Based on network pharmacology approaches, we attempted to unveil the mechanisms of FDY003, an herbal drug comprised of Lonicera japonica Thunberg, Artemisia capillaris Thunberg, and Cordyceps militaris, against BC at a systemic level. We found that FDY003 exhibited pharmacological effects on human BC cells. Subsequently, detailed data regarding the biochemical components contained in FDY003 were obtained from comprehensive herbal medicine-related databases, including TCMSP and CancerHSP. By evaluating their pharmacokinetic properties, 18 chemical compounds in FDY003 were shown to be potentially active constituents interacting with 140 BC-associated therapeutic targets to produce the pharmacological activity. Gene ontology enrichment analysis using g:Profiler indicated that the FDY003 targets were involved in the modulation of cellular processes, involving the cell proliferation, cell cycle process, and cell apoptosis. Based on a KEGG pathway enrichment analysis, we further revealed that a variety of oncogenic pathways that play key roles in the pathology of BC were significantly enriched with the therapeutic targets of FDY003; these included PI3K-Akt, MAPK, focal adhesion, FoxO, TNF, and estrogen signaling pathways. Here, we present a network-perspective of the molecular mechanisms via which herbal drugs treat BC.
Collapse
Affiliation(s)
- Ho-Sung Lee
- The Fore, 87 Ogeum-ro, Songpa-gu, Seoul 05542, Republic of Korea
- Forest Hospital, 129 Ogeum-ro, Songpa-gu, Seoul 05549, Republic of Korea
| | - In-Hee Lee
- The Fore, 87 Ogeum-ro, Songpa-gu, Seoul 05542, Republic of Korea
| | - Kyungrae Kang
- Forest Hospital, 129 Ogeum-ro, Songpa-gu, Seoul 05549, Republic of Korea
| | - Sang-In Park
- Forestheal Hospital, 173 Ogeum-ro, Songpa-gu, Seoul 05641, Republic of Korea
| | - Seung-Joon Moon
- Forest Hospital, 129 Ogeum-ro, Songpa-gu, Seoul 05549, Republic of Korea
| | - Chol Hee Lee
- Forest Hospital, 129 Ogeum-ro, Songpa-gu, Seoul 05549, Republic of Korea
| | - Dae-Yeon Lee
- The Fore, 87 Ogeum-ro, Songpa-gu, Seoul 05542, Republic of Korea
- Forest Hospital, 129 Ogeum-ro, Songpa-gu, Seoul 05549, Republic of Korea
| |
Collapse
|
19
|
Chang X, Sun D, Shi D, Wang G, Chen Y, Zhang K, Tan H, Liu J, Liu B, Ouyang L. Design, synthesis, and biological evaluation of quinazolin-4(3 H)-one derivatives co-targeting poly(ADP-ribose) polymerase-1 and bromodomain containing protein 4 for breast cancer therapy. Acta Pharm Sin B 2021; 11:156-180. [PMID: 33532187 PMCID: PMC7838034 DOI: 10.1016/j.apsb.2020.06.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/08/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
This study was aimed to design the first dual-target small-molecule inhibitor co-targeting poly (ADP-ribose) polymerase-1 (PARP1) and bromodomain containing protein 4 (BRD4), which had important cross relation in the global network of breast cancer, reflecting the synthetic lethal effect. A series of new BRD4 and PARP1 dual-target inhibitors were discovered and synthesized by fragment-based combinatorial screening and activity assays that together led to the chemical optimization. Among these compounds, 19d was selected and exhibited micromole enzymatic potencies against BRD4 and PARP1, respectively. Compound 19d was further shown to efficiently modulate the expression of BRD4 and PARP1. Subsequently, compound 19d was found to induce breast cancer cell apoptosis and stimulate cell cycle arrest at G1 phase. Following pharmacokinetic studies, compound 19d showed its antitumor activity in breast cancer susceptibility gene 1/2 (BRCA1/2) wild-type MDA-MB-468 and MCF-7 xenograft models without apparent toxicity and loss of body weight. These results together demonstrated that a highly potent dual-targeted inhibitor was successfully synthesized and indicated that co-targeting of BRD4 and PARP1 based on the concept of synthetic lethality would be a promising therapeutic strategy for breast cancer.
Collapse
Key Words
- BC, breast cancer
- BET, bromodomain and extra-terminal domain
- BRCA1/2, breast cancer susceptibility gene 1/2
- BRD4
- BRD4, bromodomain 4
- CDK4/6, cyclin-dependent kinase 4/6
- DSB, DNA double-strand break
- Dual-target inhibitor
- EGFR, epidermal growth factor receptor
- ELISA, enzyme linked immunosorbent assay
- ER, estrogen receptor
- ESI-HR-MS, high-resolution mass spectra
- FDA, U.S. Food and Drug Administration
- FITC, fluorescein isothiocyanate isomer I
- HE, hematoxylin-eosin
- HPLC, high-performance liquid chromatography
- HR, homologous recombination
- HRD, homologous recombination deficiency
- IHC, immunohistochemistry
- NHEJ, nonhomologous end-joining
- PARP1
- PARP1, poly(ADP-ribose) polymerase-1
- PI, propidium iodide
- PK, pharmacokinetics
- PPI, protein−protein interaction
- Quinazolin-4(3H)-one derivatives
- SAR, structure–activity relationship
- SOP, standard operation process
- Synthetic lethality
- TCGA, the cancer genome atlas
- TGI, tumor growth inhibition
- TLC, thin-layer chromatography
- TNBC, triple-negative breast cancer
- TR-FRET, time-resolved fluorescence resonance energy transfer.
- shRNA, short hairpin RNA
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jie Liu
- Corresponding authors. Tel./fax: +86 28 85503817 (Jie Liu), +86 28 85164063 (Bo Liu), +86 28 85503817 (Liang Ouyang).
| | - Bo Liu
- Corresponding authors. Tel./fax: +86 28 85503817 (Jie Liu), +86 28 85164063 (Bo Liu), +86 28 85503817 (Liang Ouyang).
| | - Liang Ouyang
- Corresponding authors. Tel./fax: +86 28 85503817 (Jie Liu), +86 28 85164063 (Bo Liu), +86 28 85503817 (Liang Ouyang).
| |
Collapse
|
20
|
Fang J, Sheng X, Bao H, Zhang Y, Lu H. Comparative analysis of intact glycopeptides from mannose receptor among different breast cancer subtypes using mass spectrometry. Talanta 2020; 223:121676. [PMID: 33303137 DOI: 10.1016/j.talanta.2020.121676] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 01/05/2023]
Abstract
Breast cancer is a highly heterogeneous disease, encompassing a number of biologically distinct entities with specific pathologic features and biological behaviors. In the preliminary experiments, we identified several glycosylation sites of mannose receptors in different breast cancer subtypes and showed that the mannose receptors could be a potential marker for breast cancer. However, the glycan composition on each site is still unknown because the glycan was removed by PNGase F in previous work. Analysis of intact glycopeptides can provide the information of both the glycan composition and the glycosylation site, which can further help to reveal the difference of glycosylation in the four subtypes of breast cancer. In this work, we analyzed the intact glycopeptides of the mannose receptors in serum from breast cancer patients using isobaric tags for relative and absolute quantitation (iTRAQ) and LC-MS/MS. In total, 7 glycosylation sites and 12 glycan types corresponding to 26 intact glycopeptides were characterized from the four subtypes of breast cancer. Among them, 11 glycopeptides can be used to differentiate the subtypes of breast cancer, which further supported the previous conclusion that mannose receptor can be used as a potential marker for the identification of breast cancer subtypes.
Collapse
Affiliation(s)
- Jing Fang
- Shanghai Cancer Center and Department of Chemistry, Fudan University, Shanghai, 200032, PR China; Institutes of Biomedical Sciences and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, 200433, PR China
| | - Xiangying Sheng
- Shanghai Cancer Center and Department of Chemistry, Fudan University, Shanghai, 200032, PR China
| | - Huimin Bao
- Shanghai Cancer Center and Department of Chemistry, Fudan University, Shanghai, 200032, PR China
| | - Ying Zhang
- Institutes of Biomedical Sciences and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, 200433, PR China
| | - Haojie Lu
- Shanghai Cancer Center and Department of Chemistry, Fudan University, Shanghai, 200032, PR China; Institutes of Biomedical Sciences and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, 200433, PR China.
| |
Collapse
|
21
|
Crizotinib induced antitumor activity and synergized with chemotherapy and hormonal drugs in breast cancer cells via downregulating MET and estrogen receptor levels. Invest New Drugs 2020; 39:77-88. [PMID: 32833135 DOI: 10.1007/s10637-020-00989-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 08/16/2020] [Indexed: 02/07/2023]
Abstract
MET is a receptor tyrosine kinase known to drive neoplastic transformation and aggressive tumor phenotypes. Crizotinib is an oral multi-targeted tyrosine kinase inhibitor of MET, ALK, RON, and ROS1 kinases. In this study, the anticancer effects of crizotinib on breast cancer cells were investigated in vitro along with the molecular mechanisms associated with these effects. Besides, the antiproliferative effects of crizotinib in combination with chemotherapy, hormonal drugs, and targeted agents were examined. Results showed that crizotinib produced dose-dependent antiproliferative effects in BT-474 and SK-BR-3 breast cancer cells with IC50 values of 1.7 μM and 5.2 μM, respectively. Crizotinib inhibited colony formation of BT-474 cells at low micromolar concentrations (1-5 μM). Immunofluorescence and Western blotting indicated that crizotinib reduced total levels of MET and estrogen receptor (ERα) in BT-474 cells. Also, crizotinib reduced the levels of phosphorylated (active) MET and HER2 in BT-474 cells. The combined treatment of crizotinib with doxorubicin and paclitaxel resulted in synergistic growth inhibition of BT-474 cells with combination index values of 0.46 and 0.35, respectively. Synergy was also observed with the combination of crizotinib with the hormonal drugs 4-hydroxytamoxifen and fulvestrant in BT-474 cells. Alternatively, the combination of crizotinib with lapatinib produced antagonistic antiproliferative effects in both BT-474 and SK-BR-3 cells. Collectively, these findings demonstrate the anticancer effects of crizotinib in breast cancer cells and reveal ERα as a potential therapeutic target of the drug apart from its classical kinase inhibitory activity. Crizotinib could be an appealing option in combination with chemotherapy or hormonal drugs for the management of breast cancer.
Collapse
|
22
|
Arslanturk S, Draghici S, Nguyen T. Integrated Cancer Subtyping using Heterogeneous Genome-Scale Molecular Datasets. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2020; 25:551-562. [PMID: 31797627 PMCID: PMC6933742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Vast repositories of heterogeneous data from existing sources present unique opportunities. Taken individually, each of the datasets offers solutions to important domain and source-specific questions. Collectively, they represent complementary views of related data entities with an aggregate information value often well exceeding the sum of its parts. Integration of heterogeneous data is therefore paramount to i) obtain a more unified picture and comprehensive view of the relations, ii) achieve more robust results, iii) improve the accuracy and integrity, and iv) illuminate the complex interactions among data features. In this paper, we have proposed a data integration methodology to identify subtypes of cancer using multiple data types (mRNA, methylation, microRNA and somatic variants) and different data scales that come from different platforms (microarray, sequencing, etc.). The Cancer Genome Atlas (TCGA) dataset is used to build the data integration and cancer subtyping framework. The proposed data integration and disease subtyping approach accurately identifies novel subgroups of patients with significantly different survival profiles. With current availability of vast genomics, and variant data for cancer, the proposed data integration system will better differentiate cancer and patient subtypes for risk and outcome prediction and targeted treatment planning without additional cost and precious lost time.
Collapse
Affiliation(s)
- Suzan Arslanturk
- Department of Computer Science, Wayne State University Detroit, MI 48202, USA
| | - Sorin Draghici
- Department of Computer Science, Wayne State University Detroit, MI 48202, USA
| | - Tin Nguyen
- Department of Computer Science and Engineering, University of Nevada Reno, Nevada 89557, USA
| |
Collapse
|
23
|
Sonkar K, Ayyappan V, Tressler CM, Adelaja O, Cai R, Cheng M, Glunde K. Focus on the glycerophosphocholine pathway in choline phospholipid metabolism of cancer. NMR IN BIOMEDICINE 2019; 32:e4112. [PMID: 31184789 PMCID: PMC6803034 DOI: 10.1002/nbm.4112] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/16/2019] [Accepted: 04/20/2019] [Indexed: 05/02/2023]
Abstract
Activated choline metabolism is a hallmark of carcinogenesis and tumor progression, which leads to elevated levels of phosphocholine and glycerophosphocholine in all types of cancer tested so far. Magnetic resonance spectroscopy applications have played a key role in detecting these elevated choline phospholipid metabolites. To date, the majority of cancer-related studies have focused on phosphocholine and the Kennedy pathway, which constitutes the biosynthesis pathway for membrane phosphatidylcholine. Fewer and more recent studies have reported on the importance of glycerophosphocholine in cancer. In this review article, we summarize the recent literature on glycerophosphocholine metabolism with respect to its cancer biology and its detection by magnetic resonance spectroscopy applications.
Collapse
Affiliation(s)
- Kanchan Sonkar
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vinay Ayyappan
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Caitlin M. Tressler
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Oluwatobi Adelaja
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ruoqing Cai
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Menglin Cheng
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kristine Glunde
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
24
|
In-vitro effects of the FS50 protein from salivary glands of Xenopsylla cheopis on voltage-gated sodium channel activity and motility of MDA-MB-231 human breast cancer cells. Anticancer Drugs 2019; 29:880-889. [PMID: 29912729 DOI: 10.1097/cad.0000000000000662] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Voltage-gated sodium channel activity enhances the motility and oncogene expression of metastasic cancer cells that express a neonatal alternatively spliced form of the NaV1.5 isoform. We reported previously that FS50, a salivary protein from Xenopsylla cheopis, showed inhibitory activity against the NaV1.5 channel when assayed in HEK 293T cells and antiarrhythmia effects on rats and monkeys after induction of arrhythmia by BaCl2. This study aims to identify the effect of FS50 on voltage-gated sodium channel activity and the motility of MDA-MB-231 human breast cancer cells in vitro. NaV1.5 was abnormally expressed in the highly metastatic breast cancer cell line MDA-MB-231, but not in the MCF-7 cell line. FS50 significantly inhibited sodium current, migration, and invasion in a dose-dependent manner, but had no effect on the proliferation of MDA-MB-231 cells at the working concentrations (1.5-12 μmol/l) after a long-term treatment for 48 h. Meanwhile, FS50 decreased NaV1.5 mRNA expression without altering the total protein level in MDA-MB-231 cells. Correspondingly, the results also showed that MMP-9 activity and the ratio of MMP-9 mRNA to TIMP-1 mRNA were markedly decreased by FS50. Taken together, our findings highlighted for the first time an inhibitory effect of a salivary protein from a blood-feeding arthropod on breast cancer cells through the NaV1.5 channel. Furthermore, this study provided a new candidate leading molecule against antitumor cells expressing NaV1.5.
Collapse
|
25
|
Li S, Liu Z, Fang XD, Wang XY, Fei BY. MicroRNA (miR)-597-5p Inhibits Colon Cancer Cell Migration and Invasion by Targeting FOS-Like Antigen 2 ( FOSL2). Front Oncol 2019; 9:495. [PMID: 31245295 PMCID: PMC6581747 DOI: 10.3389/fonc.2019.00495] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/24/2019] [Indexed: 12/18/2022] Open
Abstract
Our previous work had shown that FOS-like antigen 2 (FOSL2) is regulated by miR-143-5p in colorectal cancer (CRC). Given that it has been shown by others that FOSL2 is also a target of miR-597-5p in breast adenocarcinoma, the objective of the current work was to determine whether FOSL2 is regulated by miR-597-5p in CRC and the role of miR-597-5p in CRC. MiR-597-5p expression was determined in RNA obtained from 30 paired samples of colon cancer and tumor adjacent normal tissue, as well as in the LoVo (CRC cell line) and FHC (normal colonic epithelial cells) by quantitative real time polymerase chain reaction (qRT-PCR). MiR-597-5p expression was significantly downregulated in both CRC tissue and LoVo cells. Reporter assays using wild-type and miR-597-5p seed mutant FOSL2 confirmed that FOSL2 is a bona fide target of miR-597-5p. Modulating miR-597-5p expression levels in FHC and LoVo cells using antagomir and mimic, respectively, impacted expression of epithelial and mesenchymal cell markers as well as in vitro migration and invasion, without any effect on cell proliferation, showing that miR-597-5p functions as a suppressor of epithelial to mesenchymal transition. Restoration of FOSL2 expression rescued pro-metastatic functional properties of LoVo cells conforming that effect of miR-597-5p was being mediated by targeting FOSL2. Xenograft assays in athymic nude mice showed that miR-597-5p mimic did not reduce tumor incidence or growth in LoVo cells. However, using a hepatic metastasis model showed that miR-597-5p mimic can significantly prevent hepatic metastatic nodule formation as well as FOSL2 expression in these metastatic nodules. Importantly, FOSL2 mRNA and miR-597-5p expression was found to be inversely correlated in an independent cohort of 21 CRC patients Cumulatively our results show that miR-597-5p functions as a suppressor of metastatic progression in CRC by targeting FOSL2. Replenishment of miR-597-5p can be a potential therapeutic target where its expression along with FOSL2 can serve as potential diagnostic markers in CRC.
Collapse
Affiliation(s)
- Shuo Li
- Department of Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhuo Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xue-Dong Fang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiu-Ying Wang
- Medical Record Department, China Japan Union Hospital of Jilin University, Changchun, China
| | - Bing-Yuan Fei
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
26
|
Burton LJ, Hawsawi O, Sweeney J, Bowen N, Hudson T, Odero-Marah V. CCAAT-displacement protein/cut homeobox transcription factor (CUX1) represses estrogen receptor-alpha (ER-α) in triple-negative breast cancer cells and can be antagonized by muscadine grape skin extract (MSKE). PLoS One 2019; 14:e0214844. [PMID: 30964885 PMCID: PMC6460785 DOI: 10.1371/journal.pone.0214844] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 03/21/2019] [Indexed: 11/18/2022] Open
Abstract
Triple-Negative Breast Cancers (TNBCs) are the most difficult to treat subtype of breast cancer and are often associated with high nuclear expression of Snail and Cathepsin L (Cat L) protease. We have previously shown that Snail can increase Cat L expression/activity in prostate and breast cancer cells. This study investigated the role of CUX1 (a downstream substrate of Cat L) in TNBC. We showed that Cat L and CUX1 were highly expressed in TNBC patient tissue/cell lines, as compared to ER-positive samples, using cBioportal data and western blot/zymography analyses. Additionally, luciferase reporter and chromatin immunoprecipitation assays showed that CUX1 directly bound to estrogen receptor-alpha (ER-α) promoter in MDA-MB-468, a representative TNBC cell line, and that CUX1 siRNA could restore ER-α transcription and protein expression. Furthermore, Snail and CUX1 expression in various TNBC cell lines was inhibited by muscadine grape skin extract (MSKE, a natural grape product rich in anthocyanins) or Cat L inhibitor (Z-FY-CHO) leading to decreased cell invasion and migration. MSKE decreased cell viability and increased expression of apoptotic markers in MDA-MB-468 cells, with no effect on non-tumorigenic MCF10A cells. MSKE also decreased CUX1 binding to ER-α promoter and restored ER-α expression in TNBC cells, while both MSKE and CUX1 siRNA restored sensitivity to estradiol and 4-hydoxytamoxifen as shown by increased cell viability. Therefore, CUX1 activated by Snail-Cat L signaling may contribute to TNBC via ER-α repression, and may be a viable target for TNBC using natural products such as MSKE that targets cancer and not normal cells.
Collapse
Affiliation(s)
- Liza J. Burton
- Center for Cancer Research and Therapeutic Development, Department of
Biological Sciences, Clark Atlanta University, Atlanta, Georgia, United States
of America
| | - Ohuod Hawsawi
- Center for Cancer Research and Therapeutic Development, Department of
Biological Sciences, Clark Atlanta University, Atlanta, Georgia, United States
of America
| | - Janae Sweeney
- Center for Cancer Research and Therapeutic Development, Department of
Biological Sciences, Clark Atlanta University, Atlanta, Georgia, United States
of America
| | - Nathan Bowen
- Center for Cancer Research and Therapeutic Development, Department of
Biological Sciences, Clark Atlanta University, Atlanta, Georgia, United States
of America
| | - Tamaro Hudson
- Department of Medicine, Howard University, Washington, DC, United States
of America
| | - Valerie Odero-Marah
- Center for Cancer Research and Therapeutic Development, Department of
Biological Sciences, Clark Atlanta University, Atlanta, Georgia, United States
of America
| |
Collapse
|
27
|
Hsing M, Wang Y, Rennie PS, Cox ME, Cherkasov A. ETS transcription factors as emerging drug targets in cancer. Med Res Rev 2019; 40:413-430. [PMID: 30927317 DOI: 10.1002/med.21575] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/07/2019] [Accepted: 03/07/2019] [Indexed: 12/11/2022]
Abstract
The ETS family of proteins consists of 28 transcription factors, many of which have been implicated in development and progression of a variety of cancers. While one family member, ERG, has been rigorously studied in the context of prostate cancer where it plays a critical role, other ETS factors keep emerging as potential hallmark oncodrivers. In recent years, numerous studies have reported initial discoveries of small molecule inhibitors of ETS proteins and opened novel avenues for ETS-directed cancer therapies. This review summarizes the state of the art data on therapeutic targeting of ETS family members and highlights the corresponding drug discovery strategies.
Collapse
Affiliation(s)
- Michael Hsing
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Paul S Rennie
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael E Cox
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Artem Cherkasov
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
28
|
Zou H, Sevigny MB, Liu S, Madden DT, Louie MC. Novel flexible heteroarotinoid, SL-1-39, inhibits HER2-positive breast cancer cell proliferation by promoting lysosomal degradation of HER2. Cancer Lett 2019; 443:157-166. [PMID: 30503556 DOI: 10.1016/j.canlet.2018.11.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/21/2018] [Accepted: 11/24/2018] [Indexed: 10/27/2022]
Abstract
SL-1-39 [1-(4-chloro-3-methylphenyl)-3-(4-nitrophenyl)thiourea] is a new flexible heteroarotinoid (Flex-Het) analog derived from the parental compound, SHetA2, previously shown to inhibit cell growth across multiple cancer types. The current study aims to determine growth inhibitory effects of SL-1-39 across the different subtypes of breast cancer cells and delineate its molecular mechanism. Our results demonstrate that while SL-1-39 blocks cell proliferation of all breast cancer subtypes tested, it has the highest efficacy against HER2+ breast cancer cells. Molecular analyses suggest that SL-1-39 prevents S phase progression of HER2+ breast cancer cells (SKBR3 and MDA-MB-453), which is consistent with reduced expression of key cell-cycle regulators at both the protein and transcriptional levels. SL-1-39 treatment also decreases the protein levels of HER2 and pHER2 as well as its downstream effectors, pMAPK and pAKT. Reduction of HER2 and pHER2 at the protein level is attributed to increased lysosomal degradation of total HER2 levels. This is the first study to show that a flexible heteroarotinoid analog modulates the HER2 signaling pathway through lysosomal degradation, and thus further warrants the development of SL-1-39 as a therapeutic option for HER2+ breast cancer.
Collapse
Affiliation(s)
- Hongye Zou
- Department of Natural Sciences and Mathematics, Dominican University of California, 50 Acacia Avenue, San Rafael, CA, 94901, USA.
| | - Mary B Sevigny
- Department of Natural Sciences and Mathematics, Dominican University of California, 50 Acacia Avenue, San Rafael, CA, 94901, USA.
| | - Shengquan Liu
- College of Pharmacy, Touro University California, 1310 Club Drive, Vallejo, CA, 94594, USA.
| | - David T Madden
- College of Pharmacy, Touro University California, 1310 Club Drive, Vallejo, CA, 94594, USA; Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA.
| | - Maggie C Louie
- Department of Natural Sciences and Mathematics, Dominican University of California, 50 Acacia Avenue, San Rafael, CA, 94901, USA; College of Pharmacy, Touro University California, 1310 Club Drive, Vallejo, CA, 94594, USA.
| |
Collapse
|
29
|
Fang J, Tao T, Zhang Y, Lu H. A barcode mode based on glycosylation sites of membrane type mannose receptor as a new potential diagnostic marker for breast cancer. Talanta 2019; 191:21-26. [DOI: 10.1016/j.talanta.2018.08.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/28/2018] [Accepted: 08/05/2018] [Indexed: 01/08/2023]
|
30
|
Kamalabadi-Farahani M, Vasei M, Ahmadbeigi N, Ebrahimi-Barough S, Soleimani M, Roozafzoon R. Anti-tumour effects of TRAIL-expressing human placental derived mesenchymal stem cells with curcumin-loaded chitosan nanoparticles in a mice model of triple negative breast cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S1011-S1021. [DOI: 10.1080/21691401.2018.1527345] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Mohammad Kamalabadi-Farahani
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Vasei
- Department of Pathology, Molecular and Cell Biology Laboratory, Shariati Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Naser Ahmadbeigi
- Cell Based Therapies Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Tarbiat Modares University, Tehran, Iran
| | - Reza Roozafzoon
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
31
|
Kamalabadi-Farahani M, Vasei M, Ahmadbeigi N, Ebrahimi-Barough S, Soleimani M, Roozafzoon R. Anti-tumour effects of TRAIL-expressing human placental derived mesenchymal stem cells with curcumin-loaded chitosan nanoparticles in a mice model of triple negative breast cancer. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2018. [DOI: https://doi.org/10.1080/21691401.2018.1527345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Mohammad Kamalabadi-Farahani
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Vasei
- Department of Pathology, Molecular and Cell Biology Laboratory, Shariati Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Naser Ahmadbeigi
- Cell Based Therapies Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Tarbiat Modares University, Tehran, Iran
| | - Reza Roozafzoon
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
32
|
GCSH antisense regulation determines breast cancer cells' viability. Sci Rep 2018; 8:15399. [PMID: 30337557 PMCID: PMC6193953 DOI: 10.1038/s41598-018-33677-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 09/27/2018] [Indexed: 11/12/2022] Open
Abstract
Since it is known that cancer cells exhibit a preference for increased glycine consumption, the respective glycine metabolizing enzymes are in focus of many research projects. However, no cancer associated studies are available for the Glycine Cleavage System Protein H (GCSH) to date. Our initial analysis revealed a GCSH-overexpression of the protein-coding transcript variant 1 (Tv1) in breast cancer cells and tissue. Furthermore, a shorter (391 bp) transcript variant (Tv*) was amplified with an increased expression in healthy breast cells and a decreased expression in breast cancer samples. The Tv1/Tv* transcript ratio is 1.0 in healthy cells on average, and between 5–10 in breast cancer cells. Thus, a GCSH-equilibrium at the transcript level is likely conceivable for optimal glycine degradation. A possible regulative role of Tv* was proven by Tv1-Tv*-RNA-binding and overexpression studies which consequently led to serious physiological alterations: decreased metabolic activity, release of the lactate dehydrogenase, increased extracellular acidification, and finally necrosis as a result of impaired plasma membranes. In contrast, Tv1-overexpression led to an additional increase in cellular vitality of the tumor cells, primarily due to the acceleration of the mitochondrial glycine decarboxylation activity. Ultimately, we provide the first evidence of a sensitive GCSH-antisense regulation which determines cancerous cell viability.
Collapse
|
33
|
Pai S, Bader GD. Patient Similarity Networks for Precision Medicine. J Mol Biol 2018; 430:2924-2938. [PMID: 29860027 PMCID: PMC6097926 DOI: 10.1016/j.jmb.2018.05.037] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 05/24/2018] [Accepted: 05/29/2018] [Indexed: 02/08/2023]
Abstract
Clinical research and practice in the 21st century is poised to be transformed by analysis of computable electronic medical records and population-level genome-scale patient profiles. Genomic data capture genetic and environmental state, providing information on heterogeneity in disease and treatment outcome, but genomic-based clinical risk scores are limited. Achieving the goal of routine precision medicine that takes advantage of these rich genomics data will require computational methods that support heterogeneous data, have excellent predictive performance, and ideally, provide biologically interpretable results. Traditional machine-learning approaches excel at performance, but often have limited interpretability. Patient similarity networks are an emerging paradigm for precision medicine, in which patients are clustered or classified based on their similarities in various features, including genomic profiles. This strategy is analogous to standard medical diagnosis, has excellent performance, is interpretable, and can preserve patient privacy. We review new methods based on patient similarity networks, including Similarity Network Fusion for patient clustering and netDx for patient classification. While these methods are already useful, much work is required to improve their scalability for contemporary genetic cohorts, optimize parameters, and incorporate a wide range of genomics and clinical data. The coming 5 years will provide an opportunity to assess the utility of network-based algorithms for precision medicine.
Collapse
Affiliation(s)
- Shraddha Pai
- The Donnelly Centre, University of Toronto, Toronto, Canada
| | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Canada; Department of Computer Science, University of Toronto, Toronto, Canada; The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada.
| |
Collapse
|
34
|
Rautray S, Panikar S, Amutha T, Rajananthini AU. Anticancer activity of Adiantum capillus veneris and Pteris quadriureta L. in human breast cancer cell lines. Mol Biol Rep 2018; 45:1897-1911. [PMID: 30194562 DOI: 10.1007/s11033-018-4337-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 08/28/2018] [Indexed: 10/28/2022]
Abstract
Breast cancer is the most common cancer worldwide that costs lives of millions of people every year. Plant products with potential anticancer activities have become a vital source of novel agents in treating cancer. Adiantum capillus veneris (ACV) and Pteris Quadriureta (PQ) are such traditional herbs with potential pharmacological properties. In this study, both crude methanol extract and gold nanoparticles of ACV and PQ were tested for their anticancer activities in MCF7 and BT47 cell lines. By using GC-MS, we have identified 23 and 28 bioactive compounds in ACV and PQ respectively. We analysed the effects of ACV and PQ nanoparticles on various proteins involved in cell cycle and apoptosis using western blotting and PCR. With the help of flow cytometry, we measured number of cells undergoing apoptosis. We found that both the crude extract and nanoparticles have anti-proliferative and apoptosis inducing properties against MCF7 and BT47 cell lines. We also performed molecular docking to check whether there were any interactions between proteins involved in apoptosis and cell cycle and bioactive compounds present in the plant extracts. By using docking analysis, we also showed that phytol and eicosapentaenoic acid present in ACV and PQ interact with Bcl2 and cyclin D1. These findings demonstrate that ACV and PQ possess anticancer activities by modulating proteins involved in cell cycle and apoptosis. ACV and PQ that effectively modulate various oncogenic molecules can be used as promising agent for cancer therapy.
Collapse
Affiliation(s)
- Satabdi Rautray
- Department of Biotechnology, Mother Teresa Women's University, Kodaikanal, Tamil Nadu, 624101, India.
| | - Sukanya Panikar
- Department of Biotechnology, Mother Teresa Women's University, Kodaikanal, Tamil Nadu, 624101, India
| | - T Amutha
- Department of Physics, Mother Teresa Women's University, Kodaikanal, India
| | - A Usha Rajananthini
- Department of Biotechnology, Mother Teresa Women's University, Kodaikanal, Tamil Nadu, 624101, India
| |
Collapse
|
35
|
Sixou S, Müller K, Jalaguier S, Kuhn C, Harbeck N, Mayr D, Engel J, Jeschke U, Ditsch N, Cavaillès V. Importance of RIP140 and LCoR Sub-Cellular Localization for Their Association With Breast Cancer Aggressiveness and Patient Survival. Transl Oncol 2018; 11:1090-1096. [PMID: 30007204 PMCID: PMC6070698 DOI: 10.1016/j.tranon.2018.06.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/13/2018] [Accepted: 06/15/2018] [Indexed: 11/29/2022] Open
Abstract
New markers are needed to improve diagnosis and to personalize treatments for patients with breast cancer (BC). Receptor-interacting protein of 140 kDa (RIP140) and ligand-dependent corepressor (LCoR), two transcriptional co-regulators of estrogen receptors, strongly interact in BC cells. Although their role in cancer progression has been outlined in the last few years, their function in BC has not been elucidated yet. In this study, we investigated RIP140 and LCoR localization (cytoplasm vs nucleus) in BC samples from a well-characterized cohort of patients (n = 320). RIP140 and LCoR were expressed in more than 80% of tumors, (predominantly in the cytoplasm), and the two markers were highly correlated. Expression of RIP140 and LCoR in the nucleus was negatively correlated with tumor size. Conversely, RIP140 and LCoR cytoplasmic expression strongly correlated with expression of two tumor aggressiveness markers: N-cadherin and CD133 (epithelial mesenchymal transition and cancer stem cell markers, respectively). Finally, high RIP140 nuclear expression was significantly correlated with longer overall survival, whereas high total or cytoplasmic expression of RIP140 was associated with shorter disease-free survival. Our study strongly suggests that the role of RIP140 and LCoR in BC progression could vary according to their prevalent sub-cellular localization, with opposite prognostic values for nuclear and cytoplasmic expression. The involvement in BC progression/invasiveness of cytoplasmic RIP140 could be balanced by the anti-tumor action of nuclear RIP140, thus explaining the previous contradictory findings about its role in BC.
Collapse
Affiliation(s)
- Sophie Sixou
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistrasse 11, D-80337 München, Germany; Université Paul Sabatier Toulouse III, Faculté des Sciences Pharmaceutiques, F-31062 Toulouse cedex 09, France.
| | - Katharina Müller
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistrasse 11, D-80337 München, Germany.
| | - Stéphan Jalaguier
- IRCM - Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université Montpellier, Parc Euromédecine, 208 rue des Apothicaires, F-34298 Montpellier Cedex 5, France.
| | - Christina Kuhn
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistrasse 11, D-80337 München, Germany.
| | - Nadia Harbeck
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Klinikum der Ludwig-Maximilians-Universität, Maistrasse 11, D-80337 München, Germany.
| | - Doris Mayr
- Department of Pathology, Campus Innenstadt, Ludwig-Maximilians-University Hospital, Thalkirchner Str. 36, D-80337 Munich, Germany.
| | - Jutta Engel
- Tumorregister München (TRM) des Tumorzentrums München (TZM) am Klinikum der Universität München (KUM), Marchionistraße 15, 81377 Munich, Germany.
| | - Udo Jeschke
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistrasse 11, D-80337 München, Germany.
| | - Nina Ditsch
- Department of Obstetrics and Gynaecology, Campus Großhadern, Ludwig-Maximilians-University Hospital, Marchionistraße 15, 81377 Munich, Germany.
| | - Vincent Cavaillès
- IRCM - Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université Montpellier, Parc Euromédecine, 208 rue des Apothicaires, F-34298 Montpellier Cedex 5, France.
| |
Collapse
|
36
|
Roshandel D, Eslani M, Baradaran-Rafii A, Cheung AY, Kurji K, Jabbehdari S, Maiz A, Jalali S, Djalilian AR, Holland EJ. Current and emerging therapies for corneal neovascularization. Ocul Surf 2018; 16:398-414. [PMID: 29908870 DOI: 10.1016/j.jtos.2018.06.004] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 06/10/2018] [Accepted: 06/12/2018] [Indexed: 02/08/2023]
Abstract
The cornea is unique because of its complete avascularity. Corneal neovascularization (CNV) can result from a variety of etiologies including contact lens wear; corneal infections; and ocular surface diseases due to inflammation, chemical injury, and limbal stem cell deficiency. Management is focused primarily on the etiology and pathophysiology causing the CNV and involves medical and surgical options. Because inflammation is a key factor in the pathophysiology of CNV, corticosteroids and other anti-inflammatory medications remain the mainstay of treatment. Anti-VEGF therapies are gaining popularity to prevent CNV in a number of etiologies. Surgical options including vessel occlusion and ocular surface reconstruction are other options depending on etiology and response to medical therapy. Future therapies should provide more effective treatment options for the management of CNV.
Collapse
Affiliation(s)
- Danial Roshandel
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Medi Eslani
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA; Cincinnati Eye Institute, Edgewood, KY/ University of Cincinnati, Department of Ophthalmology, Cincinnati, OH, USA
| | - Alireza Baradaran-Rafii
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Albert Y Cheung
- Cincinnati Eye Institute, Edgewood, KY/ University of Cincinnati, Department of Ophthalmology, Cincinnati, OH, USA
| | - Khaliq Kurji
- Cincinnati Eye Institute, Edgewood, KY/ University of Cincinnati, Department of Ophthalmology, Cincinnati, OH, USA
| | - Sayena Jabbehdari
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Alejandra Maiz
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Setareh Jalali
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA.
| | - Edward J Holland
- Cincinnati Eye Institute, Edgewood, KY/ University of Cincinnati, Department of Ophthalmology, Cincinnati, OH, USA.
| |
Collapse
|
37
|
Yang M, Li Z, Ren M, Li S, Zhang L, Zhang X, Liu F. Stromal Infiltration of Tumor-Associated Macrophages Conferring Poor Prognosis of Patients with Basal-Like Breast Carcinoma. J Cancer 2018; 9:2308-2316. [PMID: 30026826 PMCID: PMC6036715 DOI: 10.7150/jca.25155] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 03/30/2018] [Indexed: 12/12/2022] Open
Abstract
Aims: Tumor associated macrophages (TAMs) play a critical role in the initiation and progression of breast cancer. However, their prognostic significance in the molecular subtype of basal-like breast cancer (BLBC) is poorly understood. The aim of this study was to investigate the extent and patterns of TAMs in BLBC and their associations with clinicopathological features and patient survival. Methods and Results: We evaluated TAMs in 200 cases of BLBC by immunohistochemistry using the M2 macrophage marker CD163 and the pan-macrophage marker CD68 in tumor nest and stroma, and assessed their prognostic significance. The study demonstrated that infiltration of CD163+ and CD68+ macrophages in tumor stroma was of clinical relevance in BLBC, but not those in tumor nest. Increased stromal infiltration of CD68+ or CD163+ macrophages correlated with larger tumor size, higher histological grade, higher 5-year recurrence and 5-year breast cancer mortality. Although both of CD68+ and CD163+ macrophages in tumor stroma were associated with poor recurrence-free survival (RFS) and overall survival (OS), multivariate analysis demonstrated that only CD163+ macrophage was an independent predictor of RFS and OS. Conclusions: Our results highlight the prognostic importance of TAMs' location in BLBC. CD163, a highly specific biomarker for M2 macrophages, is an independent prognostic marker for BLBC patients, and may serve as an indicator or potential target of macrophage-centred therapeutic strategies.
Collapse
Affiliation(s)
- Mu Yang
- Department of Breast Pathology and Research Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,Department of Pathology, University Medical Center of Princeton, Plainsboro, NJ 08854, USA
| | - Zhenhua Li
- Department of Breast Pathology and Research Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Meijing Ren
- Department of Breast Pathology and Research Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Shuai Li
- Department of Breast Pathology and Research Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Lanjing Zhang
- Department of Pathology, University Medical Center of Princeton, Plainsboro, NJ 08854, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.,Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA.,Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Xinmin Zhang
- Department of Pathology, Cooper University Hospital, Cooper Medical School of Rowan University, Camden, New Jersey 08103, USA
| | - Fangfang Liu
- Department of Breast Pathology and Research Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| |
Collapse
|
38
|
Turashvili G, Lightbody ED, Tyryshkin K, SenGupta SK, Elliott BE, Madarnas Y, Ghaffari A, Day A, Nicol CJB. Novel prognostic and predictive microRNA targets for triple-negative breast cancer. FASEB J 2018; 32:fj201800120R. [PMID: 29812973 DOI: 10.1096/fj.201800120r] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Triple-negative breast cancers (TNBCs) account for ∼25% of all invasive carcinomas and represent a large subset of aggressive, high-grade tumors. Despite current research focused on understanding the genetic landscape of TNBCs, reliable prognostic and predictive biomarkers remain limited. Although dysregulated microRNAs (miRNAs) have emerged as key players in many cancer types, the role of miRNAs in TNBC disease progression is unclear. We performed miRNA profiling of 51 TNBCs by next-generation sequencing to reveal differentially expressed miRNAs. A total of 228 miRNAs were identified. Three miRNAs (miR-224-5p, miR-375, and miR-205-5p) separated the tumors based on basal status. Six miRNAs (high let-7d-3p, miR-203b-5p, and miR-324-5p; low miR-30a-3p, miR-30a-5p, and miR-199a-5p) were significantly associated with decreased overall survival (OS) and 5 miRNAs (high let-7d-3p; low miR-30a-3p, miR-30a-5p, miR-30c-5p, and miR-128-3p) with decreased relapse-free survival (RFS). On multivariate analysis, high expression of let-7d-3p and low expression of miR-30a were independent predictors of decreased OS and RFS. High expression of miR-95-3p was significantly associated with decreased OS and RFS in patients treated with anthracycline-based chemotherapy. Five miRNAs (let-7d-3p, miR-30a-3p, miR-30c-5p, miR-128-3p, and miR-95-3p) were validated by quantitative RT-PCR. Our findings unveil novel prognostic and predictive miRNA targets for TNBC, including a miRNA signature that predicts patient response to anthracycline-based chemotherapy. This may improve clinical management and/or lead to the development of novel therapies.-Turashvili, G., Lightbody, E. D., Tyryshkin, K., SenGupta, S. K., Elliott, B. E., Madarnas, Y., Ghaffari, A., Day, A., Nicol, C. J. B. Novel prognostic and predictive microRNA targets for triple-negative breast cancer.
Collapse
Affiliation(s)
- Gulisa Turashvili
- Kingston Health Sciences Centre, Kingston, Ontario, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
- Department of Pathology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Elizabeth D Lightbody
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Biology and Genetics, Queen's University Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | - Kathrin Tyryshkin
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Sandip K SenGupta
- Kingston Health Sciences Centre, Kingston, Ontario, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Bruce E Elliott
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Biology and Genetics, Queen's University Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | | | - Abdi Ghaffari
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Andrew Day
- Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - Christopher J B Nicol
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Biology and Genetics, Queen's University Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
39
|
Engel N, Adamus A, Frank M, Kraft K, Kühn J, Müller P, Nebe B, Kasten A, Seitz G. First evidence of SGPL1 expression in the cell membrane silencing the extracellular S1P siren in mammary epithelial cells. PLoS One 2018; 13:e0196854. [PMID: 29718989 PMCID: PMC5931664 DOI: 10.1371/journal.pone.0196854] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 04/20/2018] [Indexed: 11/19/2022] Open
Abstract
The bioactive lipid sphingosine-1-phosphate (S1P) is a main regulator of cell survival, proliferation, motility, and platelet aggregation, and it is essential for angiogenesis and lymphocyte trafficking. In that S1P acts as a second messenger intra- and extracellularly, it might promote cancer progression. The main cause is found in the high S1P concentration in the blood, which encourage cancer cells to migrate through the endothelial barrier into the blood vessels. The irreversible degradation of S1P is solely caused by the sphingosine-1-phosphate lyase (SGPL1). SGPL1 overexpression reduces cancer cell migration and therefore silences the endogenous S1P siren, which promotes cancer cell attraction-the main reason for metastasis. Since our previous metabolomics studies revealed an increased SGPL1 activity in association with successful breast cancer cell treatment in vitro, we further investigated expression and localization of SGPL1. Expression analyses confirmed a very low SGPL1 expression in all breast cancer samples, regardless of their subtype. Additionally, we were able to prove a novel SGPL expression in the cytoplasm membrane of non-tumorigenic breast cells by fusing three independent methods. The general SGPL1 downregulation and the loss of the plasma membrane expression resulted in S1P dependent stimulation of migration in the breast cancer cell lines MCF-7 and BT-20. Not only S1P stimulated migration could be repressed by overexpressing the natural SGPL1 variant not but also more general migratory activity was significantly reduced. Here, for the first time, we report on the SGPL1 plasma membrane location in human, non-malignant breast epithelial cell lines silencing the extracellular S1P siren in vitro, and thereby regulating pivotal cellular functions. Loss of this plasma membrane distribution as well as low SGPL1 expression levels could be a potential prognostic marker and a viable target for therapy. Therefore, the precise role of SGPL1 for cancer treatment should be evaluated.
Collapse
Affiliation(s)
- Nadja Engel
- Department of Pediatric Surgery, University Hospital Marburg, Baldingerstraße, Marburg, Germany
- Department of Cell Biology, University Medicine Rostock, Schillingallee, Rostock, Germany
- Department of Oral and Maxillofacial Surgery, Facial Plastic Surgery, Rostock University Medical Center, Schillingallee, Rostock, Germany
- * E-mail: ,
| | - Anna Adamus
- Department of Pediatric Surgery, University Hospital Marburg, Baldingerstraße, Marburg, Germany
- Department of Cell Biology, University Medicine Rostock, Schillingallee, Rostock, Germany
| | - Marcus Frank
- Medical Biology and Electron Microscopy Centre, University Medicine Rostock, Strempelstraße, Rostock, Germany
| | - Karin Kraft
- Complementary Medicine, Center of Internal Medicine, University Medicine Rostock, Ernst-Heydemann-Straße, Rostock, Germany
| | - Juliane Kühn
- Department of Cell Biology, University Medicine Rostock, Schillingallee, Rostock, Germany
- Institute for Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer, Greifswald-Insel Riems, Germany
| | - Petra Müller
- Department of Cell Biology, University Medicine Rostock, Schillingallee, Rostock, Germany
| | - Barbara Nebe
- Department of Cell Biology, University Medicine Rostock, Schillingallee, Rostock, Germany
| | - Annika Kasten
- Department of Oral and Maxillofacial Surgery, Facial Plastic Surgery, Rostock University Medical Center, Schillingallee, Rostock, Germany
| | - Guido Seitz
- Department of Pediatric Surgery, University Hospital Marburg, Baldingerstraße, Marburg, Germany
| |
Collapse
|
40
|
Russell J. Management of colorectal cancer patients with brain metastases. ANZ J Surg 2018; 88:126. [DOI: 10.1111/ans.14227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 08/16/2017] [Indexed: 11/26/2022]
Affiliation(s)
- Jeremy Russell
- Department of Neurosurgery; Austin Hospital; Melbourne Victoria Australia
| |
Collapse
|
41
|
He H, Sinha I, Fan R, Haldosen LA, Yan F, Zhao C, Dahlman-Wright K. c-Jun/AP-1 overexpression reprograms ERα signaling related to tamoxifen response in ERα-positive breast cancer. Oncogene 2018; 37:2586-2600. [DOI: 10.1038/s41388-018-0165-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/02/2018] [Accepted: 01/05/2018] [Indexed: 12/20/2022]
|
42
|
Huang X, Dong H, Ting Z. Cloning, expression of a truncated HSP110 protein that augments the activities of tumor antigen-specific cytotoxic and apoptosis via tHSP110-peptide complex vaccines. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:10304-10314. [PMID: 31966365 PMCID: PMC6965763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/09/2017] [Indexed: 06/10/2023]
Abstract
The present study used a genetic engineering method to express a truncated heat shock protein 110 (tHSP110) isoform in Escherichia coli and verified its ability to bind to and present macromolecular antigens. Polymerase chain reaction (PCR) was used to obtain the truncated HSP110 gene, which was expressed in E. coli. The tHSP110 protein was non-covalently coupled to the intracellular domain (ICD) of human epidermal growth factor receptor 2 (HER2/Neu) in vitro to construct the antigen peptide complex tHSP110-ICD, which was identified by a co-immunoprecipitation assay. BALB/c mice were immunized 14-day interval for three times with the HSP110, tHSP110, HSP110-ICD, tHSP110-ICD, HSP110-P851-859 (a complex formed by full-length HSP110 with a cytotoxic T lymphocyte (CTL) epitope peptide of the Her2/neu ICD) and tHSP110-P851-859 complexes. Fourteen days after the last immunization, D2F2 cells were inoculated into BALB/c mice. The in vivo tumor volume of each group was measured every three days after cell inoculation to evaluate the immunization efficacy of the vaccine in each group. The level of the IFN-γ secreted by activated lymphocytes, the specific CTLs activity was detected. Immunohistochemical staining of bcl-2 and bax were measured on the tumor tissues of each group. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) showed that the expressed tHSP110 protein was 66 kDa in size. The non-covalent coupling of tHSP110 with ICD and peptide were confirmed by a co-immunoprecipitation assay. The in vivo tumor experiment results indicated no differences in the tumor volumes of the tHSP110-ICD and HSP110-ICD groups. In contrast, the tumor volume of the tHSP110-ICD group was significantly different compared with the tumor volume of the tHSP110-P851-859 group. After the mice immunized with tHSP110-ICD, tHSP110-P851-859 complexes, the complexes have potential immunogenicity, and can induce specific CTLs activity and apoptosis in BALB/c mice. As a tumor vaccine to inhibit in vivo tumor growth, the tHSP110 has the same ability to bind macromolecular antigens and activate tumor immune responses as full-length HSP110.
Collapse
Affiliation(s)
- Xu Huang
- Jiaxing University Medical CollegeJiaxing, Zhejiang, China
| | - Han Dong
- Jiaxing University Medical CollegeJiaxing, Zhejiang, China
| | - Zhang Ting
- College of Medical Technology, Zhejiang Chinese Medical UniversityHangzhou, China
| |
Collapse
|
43
|
Application of pharmacometrics and quantitative systems pharmacology to cancer therapy: The example of luminal a breast cancer. Pharmacol Res 2017; 124:20-33. [PMID: 28735000 DOI: 10.1016/j.phrs.2017.07.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/09/2017] [Accepted: 07/14/2017] [Indexed: 12/12/2022]
Abstract
Breast cancer (BC) is the most common cancer in women, and the second most frequent cause of cancer-related deaths in women worldwide. It is a heterogeneous disease composed of multiple subtypes with distinct morphologies and clinical implications. Quantitative systems pharmacology (QSP) is an emerging discipline bridging systems biology with pharmacokinetics (PK) and pharmacodynamics (PD) leveraging the systematic understanding of drugs' efficacy and toxicity. Despite numerous challenges in applying computational methodologies for QSP and mechanism-based PK/PD models to biological, physiological, and pharmacological data, bridging these disciplines has the potential to enhance our understanding of complex disease systems such as BC. In QSP/PK/PD models, various sources of data are combined including large, multi-scale experimental data such as -omics (i.e. genomics, transcriptomics, proteomics, and metabolomics), biomarkers (circulating and bound), PK, and PD endpoints. This offers a means for a translational application from pre-clinical mathematical models to patients, bridging the bench to bedside paradigm. Not only can these models be applied to inform and advance BC drug development, but they also could aid in optimizing combination therapies and rational dosing regimens for BC patients. Here, we review the current literature pertaining to the application of QSP and pharmacometrics-based pharmacotherapy in BC including bottom-up and top-down modeling approaches. Bottom-up modeling approaches employ mechanistic signal transduction pathways to predict the behavior of a biological system. The ones that are addressed in this review include signal transduction and homeostatic feedback modeling approaches. Alternatively, top-down modeling techniques are bioinformatics reconstruction techniques that infer static connections between molecules that make up a biological network and include (1) Bayesian networks, (2) co-expression networks, and (3) module-based approaches. This review also addresses novel techniques which utilize the principles of systems biology, synthetic lethality and tumor priming, both of which are discussed in relationship to novel drug targets and existing BC therapies. By utilizing QSP approaches, clinicians may develop a platform for improved dose individualization for subpopulation of BC patients, strengthen rationale in treatment designs, and explore mechanism elucidation for improving future treatments in BC medicine.
Collapse
|
44
|
Shen H, Li L, Yang S, Wang D, Zhou S, Chen X, Tang J. Regulatory role of tumor necrosis factor receptor-associated factor 6 in breast cancer by activating the protein kinase B/glycogen synthase kinase 3β signaling pathway. Mol Med Rep 2017. [DOI: 10.3892/mmr.2017.6782] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
45
|
Masoud V, Pagès G. Targeted therapies in breast cancer: New challenges to fight against resistance. World J Clin Oncol 2017; 8:120-134. [PMID: 28439493 PMCID: PMC5385433 DOI: 10.5306/wjco.v8.i2.120] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 09/16/2016] [Accepted: 10/17/2016] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the most common type of cancer found in women and today represents a significant challenge to public health. With the latest breakthroughs in molecular biology and immunotherapy, very specific targeted therapies have been tailored to the specific pathophysiology of different types of breast cancers. These recent developments have contributed to a more efficient and specific treatment protocol in breast cancer patients. However, the main challenge to be further investigated still remains the emergence of therapeutic resistance mechanisms, which develop soon after the onset of therapy and need urgent attention and further elucidation. What are the recent emerging molecular resistance mechanisms in breast cancer targeted therapy and what are the best strategies to apply in order to circumvent this important obstacle? The main scope of this review is to provide a thorough update of recent developments in the field and discuss future prospects for preventing resistance mechanisms in the quest to increase overall survival of patients suffering from the disease.
Collapse
|
46
|
Wong-Arce A, González-Ortega O, Rosales-Mendoza S. Plant-Made Vaccines in the Fight Against Cancer. Trends Biotechnol 2017; 35:241-256. [DOI: 10.1016/j.tibtech.2016.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 11/21/2016] [Accepted: 12/07/2016] [Indexed: 12/25/2022]
|
47
|
Adelipour M, Babaei F, Mirzababaei M, Allameh A. Correlation of micro vessel density and c-Myc expression in breast tumor of mice following mesenchymal stem cell therapy. Tissue Cell 2017; 49:315-322. [PMID: 28209368 DOI: 10.1016/j.tice.2017.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 01/11/2017] [Accepted: 01/12/2017] [Indexed: 02/07/2023]
Abstract
Stem cell therapy for degenerative diseases has been established; however there are controversies over the treatment of solid tumors with stem cell transplantation. In the present study, the anti-tumor action of mesenchymal stem cells (MSCs) has been examined in a mouse model of breast cancer with emphasize on tumor growth, angiogenesis and c-Myc expression in breast tumors. For this purpose, MSCs were isolated from bone marrow of Balb/c mice and characterized. A Balb/c mouse model of breast cancer was developed and subjected to cell therapy intra venous (I.V) or intra tumor (I.T) with MSCs. Tumor growth was measured using a digital caliber for until the end of experiment (30days). Then the mice were sacrificed and their tumors were removed and processed for histopathological examination, immunohistochemical assay of CD31 and measuring of c-Myc expression using quantitative PCR. Detection of the labeled-MSCs in tumors following injection of the cells (I.V or I.T) clearly showed the homing of MSCs into tumors. Tumor growth in case of MSC-treated mice by I.V and I.T routes was inhibited by approximately 28% and 34% respectively compared to controls. The suppression of angiogenesis was reflected in Micro Vessel Density (MVD) following I.V or I.T delivery of the MSCs. c-Myc gene expression in tumor tissues of mice treated I.V or IT with MSCs was down-regulated to 28.0% and 16.0% respectively compare to control groups. In conclusion, growth inhibition of breast tumors in mice due to MSC therapy is associated with modulation of c-Myc activation and angiogenesis markers.
Collapse
Affiliation(s)
- Maryam Adelipour
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Babaei
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammadreza Mirzababaei
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
48
|
Chimge NO, Ahmed-Alnassar S, Frenkel B. Relationship between RUNX1 and AXIN1 in ER-negative versus ER-positive Breast Cancer. Cell Cycle 2017; 16:312-318. [PMID: 28055379 DOI: 10.1080/15384101.2016.1237325] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
RUNX1 plays opposing roles in breast cancer: a tumor suppressor in estrogen receptor-positive (ER+) disease and an oncogenic role in ER-negative (ER-) tumors. Potentially mediating the former, we have recently reported that RUNX1 prevents estrogen-driven suppression of the mRNA encoding the tumor suppressor AXIN1. Accordingly, AXIN1 protein expression was diminished upon RUNX1 silencing in ER+ breast cancer cells and was positively correlated with AXIN1 protein expression across tumors with high levels of ER. Here we report the surprising observation that RUNX1 and AXIN1 proteins are strongly correlated in ER- tumors as well. However, this correlation is not attributable to regulation of AXIN1 by RUNX1 or vice versa. The unexpected correlation between RUNX1, playing an oncogenic role in ER- breast cancer, and AXIN1, a well-established tumor suppressor hub, may be related to a high ratio between the expression of variant 2 and variant 1 (v2/v1) of AXIN1 in ER- compared with ER+ breast cancer. Although both isoforms are similarly regulated by RUNX1 in estrogen-stimulated ER+ breast cancer cells, the higher v2/v1 ratio in ER- disease is expected to weaken the tumor suppressor activity of AXIN1 in these tumors.
Collapse
Affiliation(s)
- Nyam-Osor Chimge
- a Department of Medicine , Keck School of Medicine of the University of Southern California , Los Angeles , CA , USA.,b Institute for Genetic Medicine, Keck School of Medicine of the University of Southern California , Los Angeles , CA , USA
| | - Sara Ahmed-Alnassar
- b Institute for Genetic Medicine, Keck School of Medicine of the University of Southern California , Los Angeles , CA , USA.,c Department of Biochemistry and Molecular Biology , Keck School of Medicine of the University of Southern California , Los Angeles , CA , USA
| | - Baruch Frenkel
- b Institute for Genetic Medicine, Keck School of Medicine of the University of Southern California , Los Angeles , CA , USA.,c Department of Biochemistry and Molecular Biology , Keck School of Medicine of the University of Southern California , Los Angeles , CA , USA.,d Department of Orthopedic Surgery , Keck School of Medicine of the University of Southern California , Los Angeles , CA , USA
| |
Collapse
|
49
|
Muccio DD, Atigadda VR, Brouillette WJ, Bland KI, Krontiras H, Grubbs CJ. Translation of a Tissue-Selective Rexinoid, UAB30, to the Clinic for Breast Cancer Prevention. Curr Top Med Chem 2017; 17:676-695. [PMID: 27320329 PMCID: PMC9904082 DOI: 10.2174/1568026616666160617093604] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 03/25/2016] [Accepted: 03/25/2016] [Indexed: 11/22/2022]
Abstract
This review focuses on our efforts to translate a low-toxicity retinoid X receptor-selective agonist, UAB30, to the clinic for the prevention of breast cancers. The review is divided into several sections. First, the current status of breast cancer prevention is discussed. Next, preclinical studies are presented that support translation of rexinoids to the clinic for cancer prevention. While current FDAapproved retinoids and rexinoids demonstrate profound effects in treating cancers, they lack sufficient safety for long term use in the high risk population that is otherwise disease free. The review stresses the need to identify cancer preventive drugs that are effective and safe in order to gain wide use in the clinic. Due to the heterogeneity of the disease, UAB30 is evaluated for the prevention of ER-positive and ER-negative mammary cancers. Since selective estrogen receptor modulators and aromatase inhibitors are used clinically to prevent and treat ER-positive breast cancers, preclinical studies also must demonstrate efficacy of UAB30 in combination with existing drugs under use in the clinic. To support an Investigational New Drug Application to the FDA, data on pharmacology and toxicity as well as mutagenicity is gathered prior to human trials. The review concludes with a discussion of the outcomes of human Phase 0/1 clinical trials that determine the safety and pharmacology of UAB30. These studies are essential before this agent is evaluated for efficacy in phase 2 trials. Success in phase 2 evaluation is critical before long-term and costly phase 3 trials are undertaken. The lack of surrogate biomarkers as endpoints for phase 2 evaluation of rexinoid preventive agents is discussed.
Collapse
Affiliation(s)
- Donald D. Muccio
- Department of Chemistry, University of Alabama at Birmingham, Birmingham Alabama 35294 USA
| | - Venkatram R Atigadda
- Department of Chemistry, University of Alabama at Birmingham, Birmingham Alabama 35294 USA
| | - Wayne J Brouillette
- Department of Chemistry, University of Alabama at Birmingham, Birmingham Alabama 35294 USA
| | - Kirby I Bland
- Department of Surgery, University of Alabama at Birmingham, Birmingham Alabama 35294 USA
| | - Helen Krontiras
- Department of Surgery, University of Alabama at Birmingham, Birmingham Alabama 35294 USA
| | - Clinton J Grubbs
- Department of Surgery, University of Alabama at Birmingham, Birmingham Alabama 35294 USA
| |
Collapse
|
50
|
Zhang X, Harbeck N, Jeschke U, Doisneau-Sixou S. Influence of vitamin D signaling on hormone receptor status and HER2 expression in breast cancer. J Cancer Res Clin Oncol 2016; 143:1107-1122. [PMID: 28025696 DOI: 10.1007/s00432-016-2325-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/16/2016] [Indexed: 12/14/2022]
Abstract
PURPOSE Breast cancer is a significant global public health issue. It is the leading cause of death among women around the world, with an incidence increasing annually. In recent years, there has been more and more information in the literature regarding a protective role of vitamin D in cancer. Increasingly preclinical and clinical studies suggest that vitamin D optimal levels can reduce the risk of breast cancer development and regulate cancer-related pathways. METHOD In this review, we focus on the importance of vitamin D in breast cancers, discussing especially the influence of vitamin D signaling on estrogen receptor and human epidermal growth factor receptor 2 (HER2), two major biomarkers of breast cancer today. CONCLUSION We discuss the possibility of actual and future targeted therapeutic approaches for vitamin D signaling in breast cancer.
Collapse
Affiliation(s)
- Xi Zhang
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany
| | - Nadia Harbeck
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany
| | - Udo Jeschke
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany
| | - Sophie Doisneau-Sixou
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany. .,Faculté des Sciences Pharmaceutiques, Université Paul Sabatier Toulouse III, 31062, Toulouse Cedex 09, France.
| |
Collapse
|