1
|
Barbieri E, Venturelli M, Mastrodomenico L, Piombino C, Ponzoni O, Zaniboni S, Barban S, Razzaboni E, Grandi G, Dominici M, Cortesi L, Toss A. Chemoprevention strategies in hereditary breast and ovarian cancer syndromes. TUMORI JOURNAL 2024:3008916241274721. [PMID: 39568367 DOI: 10.1177/03008916241274721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Hereditary breast and/or ovarian cancer syndromes are inherited disorders in which there is an increased risk of developing breast and/or ovarian cancer in the lifetime, usually at a younger age compared to the general population. Cancer prevention in these syndromes includes prophylactic surgeries, personalized surveillance programs and chemopreventive strategies. Chemoprevention exploits the use of certain drugs or other substances to help lower the risk of developing cancer. In this context, tamoxifen was the first agent considered for breast cancer prevention, followed by raloxifene and the third-generation aromatase inhibitors. On the other hand, the first and most widespread type of chemoprevention for ovarian cancer was combined hormonal contraceptive use. Although several strategies have been studied and showed promising results, only a few of these are currently applied in daily clinical practice. Side effects along with several psychological variables such as cancer perceived risk, worries and related distress, strongly influence women's decision on chemoprevention. The present review explores and summarizes the available evidence on breast and ovarian cancer chemoprevention approaches.
Collapse
Affiliation(s)
- Elena Barbieri
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Marta Venturelli
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Luciana Mastrodomenico
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Claudia Piombino
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Ornella Ponzoni
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Silvia Zaniboni
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Serena Barban
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Elisabetta Razzaboni
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Giovanni Grandi
- Obstetrics and Gynecology, Obstetrics and Gynecology Unit, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Cortesi
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Angela Toss
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
2
|
Cucciniello L, Garufi G, Di Rienzo R, Martinelli C, Pavone G, Giuliano M, Arpino G, Montemurro F, Del Mastro L, De Laurentiis M, Puglisi F. Estrogen deprivation effects of endocrine therapy in breast cancer patients: Incidence, management and outcome. Cancer Treat Rev 2023; 120:102624. [PMID: 37751658 DOI: 10.1016/j.ctrv.2023.102624] [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: 06/23/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023]
Abstract
Endocrine therapy is one of the standard adjuvant treatments to reduce the risk of recurrence and mortality in patients with hormone receptor positive early breast cancer. Despite its proven efficacy, ET side effects, which persist over time even if low grade, may deteriorate quality of life. During follow-up visits, emphasis is generally placed on the risk of disease recurrence, while the topic of ET side effects is commonly neglected and discussed only briefly. This could lead to poor adherence to therapy and early treatment discontinuation, resulting in worse survival outcomes. The aim of this review is to provide an overview of the available evidence on the incidence and reporting of ET-related side effects (including vasomotor symptoms, musculoskeletal disorders and genitourinary syndrome of menopause, as well as fatigue, psychological and ocular disorders, dysmetabolic effects and loss of bone density) and of the pharmacological and non-pharmacological strategies available to mitigate symptom burden.
Collapse
Affiliation(s)
- Linda Cucciniello
- Department of Medicine, University of Udine, Udine, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy.
| | - Giovanna Garufi
- Department of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy.
| | - Rossana Di Rienzo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Claudia Martinelli
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Giuliana Pavone
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy; Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico - San Marco", Catania, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy.
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | | | - Lucia Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy; Breast Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | | | - Fabio Puglisi
- Department of Medicine, University of Udine, Udine, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy.
| |
Collapse
|
3
|
Kumar T, Dutta RR, Thakre S, Singh A, Velagala VR, Shinde RK. Resistance to Resilience: Understanding Post-surgical Hormone Therapy in Breast Cancer Care. Cureus 2023; 15:e47869. [PMID: 38021507 PMCID: PMC10681032 DOI: 10.7759/cureus.47869] [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: 10/11/2023] [Accepted: 10/28/2023] [Indexed: 12/01/2023] Open
Abstract
Breast cancer is one of the most common types of cancer affecting women worldwide. Over the years, breast cancer has become a major public health concern, and its incidence is rising globally. The treatment of breast cancer does not stop with surgical intervention, but adjuvant therapies are administered to improve patient outcomes post-surgery based on the type of breast cancer diagnosed. This review focuses on the value of hormone therapy (HT) in improving the prognosis of breast cancer patients and why adhering to adjuvant treatment post-surgery is difficult for patients. HT aims to reduce the chances of breast cancer recurrence after surgical treatment. Even though HT is life-saving, patients tend to not adhere to the therapy due to various factors such as side effects, age-related issues, and socioeconomic status. Most patients stop adhering to the therapy as the duration can be as long as 5-10 years, and the quality of life is greatly impacted due to the side effects of the treatment. This review examines the possible factors leading to non-adherence to HT and tries to propose possible interventions that might improve patient compliance with the treatment. This article not only focuses on the impact of side effects of HT on patients' quality of life but also tries to understand the problems faced by breast cancer patients in adhering to HT.
Collapse
Affiliation(s)
- Tanishq Kumar
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Rajoshee R Dutta
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Swedaj Thakre
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Arihant Singh
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Vivek R Velagala
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Raju K Shinde
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
4
|
Abstract
Most women worldwide experience menopausal symptoms during the menopause transition or postmenopause. Vasomotor symptoms are most pronounced during the first four to seven years but can persist for more than a decade, and genitourinary symptoms tend to be progressive. Although the hallmark symptoms are hot flashes, night sweats, disrupted sleep, and genitourinary discomfort, other common symptoms and conditions are mood fluctuations, cognitive changes, low sexual desire, bone loss, increase in abdominal fat, and adverse changes in metabolic health. These symptoms and signs can occur in any combination or sequence, and the link to menopause may even be elusive. Estrogen based hormonal therapies are the most effective treatments for many of the symptoms and, in the absence of contraindications to treatment, have a generally favorable benefit:risk ratio for women below age 60 and within 10 years of the onset of menopause. Non-hormonal treatment options are also available. Although a symptom driven treatment approach with individualized decision making can improve health and quality of life for midlife women, menopausal symptoms remain substantially undertreated by healthcare providers.
Collapse
Affiliation(s)
- Erin R Duralde
- Division of Women's Health, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Talia H Sobel
- Division of Women's Health Internal Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - JoAnn E Manson
- Harvard Medical School, Boston, MA, USA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
5
|
Michael P, Roversi G, Brown K, Sharifi N. Adrenal Steroids and Resistance to Hormonal Blockade of Prostate and Breast Cancer. Endocrinology 2023; 164:bqac218. [PMID: 36580423 PMCID: PMC10091490 DOI: 10.1210/endocr/bqac218] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 12/30/2022]
Abstract
Prostate cancer and breast cancer are sex-steroid-dependent diseases that are driven in major part by gonadal sex steroids. Testosterone (T) is converted to 5α-dihydrotestosterone, both of which stimulate the androgen receptor (AR) and prostate cancer progression. Estradiol is the major stimulus for estrogen receptor-α (ERα) and proliferation of ERα-expressing breast cancer. However, the human adrenal provides an alternative source for sex steroids. A number of different androgens are produced by the adrenals, the most abundant of which is dehydroepiandrosterone (DHEA) and DHEA sulfate. These precursor steroids are subject to metabolism by peripherally expressed enzymes that are responsible for the synthesis of potent androgens and estrogens. In the case of prostate cancer, the regulation of one of these enzymatic steps occurs at least in part by way of a germline-encoded missense in 3β-hydroxysteroid dehydrogenase-1 (3βHSD1), which regulates potent androgen biosynthesis and clinical outcomes in men with advanced prostate cancer treated with gonadal T deprivation. The sex steroids that drive prostate cancer and breast cancer require a common set of enzymes for their generation. However, the pathways diverge once 3-keto, Δ4-androgens are generated and these steroids are either turned into potent androgens by steroid-5α-reductase, or into estrogens by aromatase. Alternative steroid receptors have also emerged as disease- and treatment-resistance modifiers, including a role for AR in breast cancer and glucocorticoid receptor both in breast and prostate cancer. In this review, we integrate the commonalities of adrenal steroid physiology that regulate both prostate and breast cancer while recognizing the clear distinctions between these diseases.
Collapse
Affiliation(s)
- Patrick Michael
- Genitourinary Malignancies Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Gustavo Roversi
- Genitourinary Malignancies Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Kristy Brown
- Sandra and Edward Meyer Cancer Center and Department of Medicine, Weill Cornell Medicine, New York, New York 10065, USA
| | - Nima Sharifi
- Genitourinary Malignancies Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| |
Collapse
|
6
|
Hu Y, Tse TJ, Shim YY, Purdy SK, Kim YJ, Meda V, Reaney MJT. A review of flaxseed lignan and the extraction and refinement of secoisolariciresinol diglucoside. Crit Rev Food Sci Nutr 2022; 64:5057-5072. [PMID: 36448088 DOI: 10.1080/10408398.2022.2148627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Lignan is a class of diphenolic compounds that arise from the condensation of two phenylpropanoid moieties. Oilseed and cereal crops (e.g., flaxseed, sesame seed, wheat, barley, oats, rye, etc.) are major sources of plant lignan. Methods for commercial isolation of the lignan secoisolariciresinol diglucoside (SDG) are not well reported, as most publications describing the detection, extraction, and enrichment of SDG use methods that have not been optimized for commercial scale lignan recovery. Simply scaling up laboratory methods would require expensive infrastructure to achieve a marketable yield and reproducible product quality. Therefore, establishing standard protocols to produce SDG and its derivatives on an industrial scale is critical to decrease lignan cost and increase market opportunities. This review summarizes the human health benefits of flaxseed lignan consumption, lignan physicochemical properties, and mammalian lignan metabolism, and describes methods for detecting, extracting, and enriching flaxseed lignan. Refining and optimization of these methods could lead to the development of inexpensive lignan sources for application as an ingredient in medicines, dietary supplements, and other healthy ingredients.
Collapse
Affiliation(s)
- Yingxue Hu
- Department of Chemical and Biological Engineering, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Timothy J Tse
- Department of Food and Bioproduct Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Youn Young Shim
- Department of Food and Bioproduct Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Korea
- Guangdong Saskatchewan Oilseed Joint Laboratory, Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong, China
| | - Sarah K Purdy
- Department of Food and Bioproduct Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Young Jun Kim
- Department of Food and Biotechnology, Korea University, Sejong, Korea
| | - Venkatesh Meda
- Department of Chemical and Biological Engineering, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Martin J T Reaney
- Department of Food and Bioproduct Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Guangdong Saskatchewan Oilseed Joint Laboratory, Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Allegretti M, Cesta MC, Zippoli M, Beccari A, Talarico C, Mantelli F, Bucci EM, Scorzolini L, Nicastri E. Repurposing the estrogen receptor modulator raloxifene to treat SARS-CoV-2 infection. Cell Death Differ 2022; 29:156-166. [PMID: 34404919 PMCID: PMC8370058 DOI: 10.1038/s41418-021-00844-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 07/09/2021] [Accepted: 07/25/2021] [Indexed: 12/15/2022] Open
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) necessitates strategies to identify prophylactic and therapeutic drug candidates to enter rapid clinical development. This is particularly true, given the uncertainty about the endurance of the immune memory induced by both previous infections or vaccines, and given the fact that the eradication of SARS-CoV-2 might be challenging to reach, given the attack rate of the virus, which would require unusually high protection by a vaccine. Here, we show how raloxifene, a selective estrogen receptor modulator with anti-inflammatory and antiviral properties, emerges as an attractive candidate entering clinical trials to test its efficacy in early-stage treatment COVID-19 patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Enrico M Bucci
- Sbarro Health Research Organization, Biology Department CFT, Temple University, Philadelphia, PA, USA
| | - Laura Scorzolini
- Lazzaro Spallanzani National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Emanuele Nicastri
- Lazzaro Spallanzani National Institute for Infectious Diseases, IRCCS, Rome, Italy
| |
Collapse
|
8
|
Das P, Gupta A, Desai KV. JMJD6 orchestrates a transcriptional program in favor of endocrine resistance in ER+ breast cancer cells. Front Endocrinol (Lausanne) 2022; 13:1028616. [PMID: 36419768 PMCID: PMC9678079 DOI: 10.3389/fendo.2022.1028616] [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: 08/26/2022] [Accepted: 10/14/2022] [Indexed: 11/09/2022] Open
Abstract
High expression of Jumonji domain containing protein 6 (JMJD6) is strongly associated with poor prognosis in estrogen receptor positive (ER+) breast cancer. We overexpressed JMJD6 in MCF7 cells (JOE cells) and performed RNA-seq analysis. 76% of differentially expressed genes (DEGs) overlapped with ER target genes. Pathway analysis revealed that JMJD6 upregulated a larger subset of genes related to cell proliferation as compared to ER. Interestingly, JOE cells showed a decrease in ER target gene expression prompting us to check ER levels. Indeed, JOE cells showed a significant decrease in both ESR1 and ER levels and JMJD6 siRNA transfection increased the expression of both. Additionally, JOE cells showed increased RET and ERK1 expression, events associated with resistance to endocrine therapy. Accordingly, JOE cells displayed lower sensitivity and survived better at higher doses of 4-hydroxy tamoxifen (Tam) as compared to parental MCF-7 cells. Conversely, LTED-I and TAM R that resist Tam induced death, showed high expression of JMJD6. Further, JMJD6 siRNA treatment decreased growth and improved Tam sensitivity in TAM R. Comparison of JOE DEGs with known Tam signature genes showed a substantial overlap. Overall, these data suggest that blocking ER alone in patients may not eradicate proliferation of JMJD6 expressing ER+ cells and JMJD6 may predispose and sustain endocrine therapy resistance. We propose that immunostaining for JMJD6 could be developed as a potential marker for predicting endocrine therapy resistance. Further, antagonizing JMJD6 action in women expressing higher amounts of this protein, may offer a greater clinical benefit than endocrine therapy.
Collapse
|
9
|
Akhavan Zanjani M, Rahmani S, Mehranfar S, Zarrin M, Bazyar H, Moradi Poodeh B, Zare Javid A, Hosseini SA, Sadeghian M. Soy Foods and the Risk of Fracture: A Systematic Review of Prospective Cohort Studies. Complement Med Res 2021; 29:172-181. [PMID: 34547749 DOI: 10.1159/000519036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 08/16/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVES The primary objective of our study was to systematically review all available prospective cohort studies which investigated the association of soy food intake and incident fracture risk. METHODS We searched PubMed, Scopus, and Embase databases for relevant studies up to June 2021. SYNTHESIS Of 695 records, a total of 5 cohort studies were included in the current systematic review. Two studies that were performed in China evaluated hip fracture while 2 studies that were done in Singapore evaluated any kind of fractures. The other study was conducted in Japan and evaluated osteoporosis fractures. All studies used a face-to-face interview to assess the dietary intake of soy foods. All 5 cohort studies were determined to be of high quality. One study considered soy food as a part of a vegetables-fruit-soy food dietary pattern. Others reported the association of dietary intake of soy foods with the risk of fractures. CONCLUSION The evidence from prospective cohort studies was suggestive for a protective role of soy foods, alone or within a dietary pattern, in the risk of incident fracture among Asian women, particularly for those in early menopause and those who used fermented soy products. But for men, the association was not significant. However, more cohort studies, including non-Asian populations, are required to confirm this association fully.
Collapse
Affiliation(s)
- Mohsen Akhavan Zanjani
- Division of Kinesiology, School of Health and Human Performance, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Sepideh Rahmani
- Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sanaz Mehranfar
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, University of Medical Sciences, Tehran, Iran
| | - Milad Zarrin
- Musculoskeletal Rehabilitation Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hadi Bazyar
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bahman Moradi Poodeh
- Department of Laboratory, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Ahmad Zare Javid
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Ahmad Hosseini
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mehdi Sadeghian
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| |
Collapse
|
10
|
Shen YT, Huang X, Zhang G, Jiang B, Li CJ, Wu ZS. Pan-Cancer Prognostic Role and Targeting Potential of the Estrogen-Progesterone Axis. Front Oncol 2021; 11:636365. [PMID: 34322374 PMCID: PMC8311599 DOI: 10.3389/fonc.2021.636365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 06/23/2021] [Indexed: 12/21/2022] Open
Abstract
Introduction Estrogen receptors (ESRs) and progesterone receptors (PGRs) are associated with the development and progression of various tumors. The feasibility of ESRs and PGRs as prognostic markers and therapeutic targets for multiple cancers was evaluated via pan-cancer analysis. Methods The pan-cancer mRNA expression levels, genetic variations, and prognostic values of ESR1, ESR2, and PGR were analyzed using the Gene Expression Profiling Interactive Analysis 2 (GEPIA2) and cBioPortal. The expression levels of ERa, ERb, and PGR proteins were detected by immunohistochemical staining using paraffin-embedded tissue specimens of ovarian serous cystadenocarcinoma (OV) and uterine endometrioid adenocarcinoma (UTEA). Correlation between immunomodulators and immune cells was determined based on the Tumor and Immune System Interaction Database (TISIDB). Results ESR1, ESR2, and PGR mRNAs were found to be differentially expressed in different cancer types, and were associated with tumor progression and clinical prognosis. ERa, ERb, and PGR proteins were further determined to be significantly differentially expressed in OV and UTEA via immunohistochemical staining. The expression of ERa protein was positively correlated with a high tumor stage, whereas the expression of PGR protein was conversely associated with a high tumor stage in patients with OV. In patients with UTEA, the expression levels of both ERa and PGR proteins were conversely associated with tumor grade and stage. In addition, the expression levels of ESR1, ESR2, and PGR mRNAs were significantly associated with the expression of immunomodulators and immune cells. Conclusion ESR1, ESR2, and PGR are potential prognostic markers and therapeutic targets, as well as important factors for the prediction, evaluation, and individualized treatment in several cancer types.
Collapse
Affiliation(s)
- Yu-Ting Shen
- Department of Pathology, Anhui Medical University, Hefei, China.,Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,The Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Gang Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bo Jiang
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Cheng-Jun Li
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Zheng-Sheng Wu
- Department of Pathology, Anhui Medical University, Hefei, China.,Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
11
|
Masaret GS. Convenient synthesis and anticancer evaluation of novel pyrazolyl‐thiophene, thieno[3,2‐
b
]pyridine, pyrazolo[3,4‐
d
]thieno[3,2‐
b
]pyridine and pyrano[2,3‐
d
]thieno[3,2‐
b
]pyridine derivatives. J Heterocycl Chem 2021. [DOI: 10.1002/jhet.4262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Ghada S. Masaret
- Chemistry Department College of Applied Sciences, Umm Al‐Qura University Makkah Saudi Arabia
| |
Collapse
|
12
|
Panossian AG, Efferth T, Shikov AN, Pozharitskaya ON, Kuchta K, Mukherjee PK, Banerjee S, Heinrich M, Wu W, Guo D, Wagner H. Evolution of the adaptogenic concept from traditional use to medical systems: Pharmacology of stress- and aging-related diseases. Med Res Rev 2021; 41:630-703. [PMID: 33103257 PMCID: PMC7756641 DOI: 10.1002/med.21743] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 08/26/2020] [Accepted: 10/11/2020] [Indexed: 12/14/2022]
Abstract
Adaptogens comprise a category of herbal medicinal and nutritional products promoting adaptability, resilience, and survival of living organisms in stress. The aim of this review was to summarize the growing knowledge about common adaptogenic plants used in various traditional medical systems (TMS) and conventional medicine and to provide a modern rationale for their use in the treatment of stress-induced and aging-related disorders. Adaptogens have pharmacologically pleiotropic effects on the neuroendocrine-immune system, which explain their traditional use for the treatment of a wide range of conditions. They exhibit a biphasic dose-effect response: at low doses they function as mild stress-mimetics, which activate the adaptive stress-response signaling pathways to cope with severe stress. That is in line with their traditional use for preventing premature aging and to maintain good health and vitality. However, the potential of adaptogens remains poorly explored. Treatment of stress and aging-related diseases require novel approaches. Some combinations of adaptogenic plants provide unique effects due to their synergistic interactions in organisms not obtainable by any ingredient independently. Further progress in this field needs to focus on discovering new combinations of adaptogens based on traditional medical concepts. Robust and rigorous approaches including network pharmacology and systems pharmacology could help in analyzing potential synergistic effects and, more broadly, future uses of adaptogens. In conclusion, the evolution of the adaptogenic concept has led back to basics of TMS and a new level of understanding of holistic approach. It provides a rationale for their use in stress-induced and aging-related diseases.
Collapse
Affiliation(s)
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and BiochemistryJohannes Gutenberg UniversityMainzGermany
| | - Alexander N. Shikov
- Department of technology of dosage formsSaint‐Petersburg State Chemical‐Pharmaceutical UniversitySt. PetersburgRussia
| | - Olga N. Pozharitskaya
- Department of BiotechnologyMurmansk Marine Biological Institute of the Kola Science Center of the Russian Academy of Sciences (MMBI KSC RAS)MurmanskRussia
| | - Kenny Kuchta
- Department of Far Eastern Medicine, Clinic for Gastroenterology and Gastrointestinal OncologyUniversity Medical Center GöttingenGöttingenGermany
| | - Pulok K. Mukherjee
- Department of Pharmaceutical Technology, School of Natural Product StudiesJadavpur UniversityKolkataIndia
| | - Subhadip Banerjee
- Department of Pharmaceutical Technology, School of Natural Product StudiesJadavpur UniversityKolkataIndia
| | - Michael Heinrich
- Research Cluster Biodiversity and Medicines, UCL School of Pharmacy, Centre for Pharmacognosy and PhytotherapyUniversity of LondonLondonUK
| | - Wanying Wu
- Shanghai Research Center for TCM Modernization, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| | - De‐an Guo
- Shanghai Research Center for TCM Modernization, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| | - Hildebert Wagner
- Department of Pharmacy, Center for Pharma ResearchLudwig‐Maximilians‐Universität MünchenMunichGermany
| |
Collapse
|
13
|
AlOmeir O, Patel N, Donyai P. Adherence to adjuvant endocrine therapy among breast cancer survivors: a systematic review and meta-synthesis of the qualitative literature using grounded theory. Support Care Cancer 2020; 28:5075-5084. [PMID: 32601852 PMCID: PMC7546985 DOI: 10.1007/s00520-020-05585-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 06/18/2020] [Indexed: 11/26/2022]
Abstract
PURPOSE Numerous studies have examined non-adherence to adjuvant endocrine therapy in women recovering from breast cancer, but none provides a comprehensive theory to explain the challenges of long-term medication taking and resilience needed to continue. The aim of this study was to source, appraise, and synthesize data from existing qualitative studies to develop an in-depth explanatory model of non-adherence and discontinuation of hormonal medication among breast cancer survivors. METHODS A comprehensive search of databases and the literature identified 24 eligible qualitative studies published 2010-2019. Quotations (n = 801) listed within these papers and the original author interpretations were synthesized using NVivo, and grounded theory methodology. RESULTS At the beginning, knowledge about adjuvant endocrine therapy, trust in doctors, and worries and expectations, mean agreeing to medication is the only viable option, akin to a Hobson's choice. Thereafter, women's ability to deal with medication side-effects, knowledge and support received affect their decision to continue, akin to a horned dilemma where giving up the medication risks cancer recurrence and continuing means reduced contentment. Women stopping medication altogether question treatment necessity, search for normalcy and prioritize quality of life. CONCLUSION Shared experiences and understandings were uncovered by examining commonalities in existing publications. The core category explained the difficulties women face with the initial decision to accept long-term endocrine therapy and then the everyday challenges of continuing or deciding to stop treatment early. An educational tool to inform survivors and health professionals about these challenges could potentially improve women's experience on treatment and in turn their adherence.
Collapse
Affiliation(s)
- Othman AlOmeir
- School of Pharmacy, University of Reading, Whiteknights Campus, Reading, UK.
- Department of Pharmacy, University of Reading, PO Box 226, Whiteknights, Reading, Berkshire, RG6 6AP, UK.
| | - Nilesh Patel
- Department of Pharmacy, University of Reading, PO Box 226, Whiteknights, Reading, Berkshire, RG6 6AP, UK
| | - Parastou Donyai
- Department of Pharmacy, University of Reading, PO Box 226, Whiteknights, Reading, Berkshire, RG6 6AP, UK
| |
Collapse
|
14
|
Abstract
Patients with a current diagnosis of breast cancer are enjoying dramatic cure rates and survivorship secondary to an increase in awareness, earlier detection, and more effective therapies. Although strategies such as Breast Cancer Awareness Month in October focus on early detection, lifestyle changes are seldom discussed other than dietary concerns and physical activity. Lifestyle modifications centered on diet and exercise have been demonstrated to affect overall disease-free survival in breast cancer. Since the early 2000s, the role of the human gut microbiota and its relation to breast cancer has become a major area of interest in the scientific and medical community. We live and survive owing to the symbiotic relationship with the microorganisms within us: the human microbiota. Scientific advances have identified a subset of the gut microbiota: the estrobolome, those bacteria that have the genetic capability to metabolize estrogen, which plays a key role in most breast cancers. Recent research provides evidence that the gut microbiome plays a substantial role in estrogen regulation. Gut microbiota diversity appears to be an essential component of overall health, including breast health. Future research attention should include a more extensive focus on the role of the human gut microbiota in breast cancer.
Collapse
Affiliation(s)
- Balazs I Bodai
- The Breast Cancer Survivorship Institute, Kaiser Permanente, Sacramento, CA
| | - Therese E Nakata
- The Breast Cancer Survivorship Institute, Kaiser Permanente, Sacramento, CA
| |
Collapse
|
15
|
Zheng N, Hsieh E, Cai H, Shi L, Gu K, Zheng Y, Bao PP, Shu XO. Soy Food Consumption, Exercise, and Body Mass Index and Osteoporotic Fracture Risk Among Breast Cancer Survivors: The Shanghai Breast Cancer Survival Study. JNCI Cancer Spectr 2019; 3:pkz017. [PMID: 31157323 PMCID: PMC6527440 DOI: 10.1093/jncics/pkz017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 01/31/2019] [Accepted: 03/18/2019] [Indexed: 11/25/2022] Open
Abstract
Background Breast cancer survivors have a high incidence of osteoporosis-related fractures; the associated factors are understudied. We investigated incidence of bone fracture and its associations with soy food consumption, exercise, and body mass index among breast cancer survivors. Methods This prospective study included 4139 stage 0–III breast cancer patients and 1987 pre-/perimenopausal and 2152 postmenopausal patients. Fractures were assessed at 18 months and at 3, 5, and 10 years after cancer diagnosis. Osteoporotic fractures were defined as fractures caused by falls from standing height and at sites associated with osteoporosis. Exercise and soy isoflavone intake were assessed at 6 and 18 months postdiagnosis. Weight and height were measured at baseline. Lifetable and Cox regression analyses were employed. All statistical tests were two sided. Results The 10-year incidence for osteoporotic fractures was 2.9% and 4.4% for pre-/perimenopausal and postmenopausal patients, respectively. High soy isoflavone intake was associated with reduced risk among pre-/perimenopausal patients (hazard ratio [HR] = 0.22, 95% confidence interval [CI] = 0.09 to 0.53, for soy isoflavone mg/d ≥56.06 vs <31.31; Ptrend < .001) but not among postmenopausal patients (Pinteraction < .01). Overweight (vs normal weight) was a risk factor for pre-/perimenopausal patients (HR = 1.81, 95% CI = 1.04 to 3.14) but not for postmenopausal patients (HR = 0.67, 95% CI = 0.43 to 1.03; Pinteraction = .01). Exercise was inversely associated with osteoporotic fractures in postmenopausal patients (HR = 0.56, 95% CI = 0.33 to 0.97, for metabolic equivalents hours ≥12.6 vs <4.5) following a dose-response pattern (Ptrend = .035), an association not modified by menopausal status. Conclusions Our findings, especially the novel association of soy food intake with osteoporotic fractures in breast cancer survivors, if confirmed, can help guide future strategies for fracture risk reduction in this vulnerable population.
Collapse
Affiliation(s)
- Neil Zheng
- Yale College, Yale University, New Haven, CT
| | - Evelyn Hsieh
- Section of Rheumatology, Yale School of Medicine, Yale University, New Haven, CT
| | - Hui Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Liang Shi
- Shanghai Municipal Center for Disease Prevention and Control, Shanghai, China
| | - Kai Gu
- Shanghai Municipal Center for Disease Prevention and Control, Shanghai, China
| | - Ying Zheng
- Shanghai Municipal Center for Disease Prevention and Control, Shanghai, China.,Shanghai Cancer Hospital, Fudan University, Shanghai, China
| | - Ping-Ping Bao
- Shanghai Municipal Center for Disease Prevention and Control, Shanghai, China
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
16
|
Lima Costa AH, Clemente WS, Bezerra KS, Lima Neto JX, Albuquerque EL, Fulco UL. Computational biochemical investigation of the binding energy interactions between an estrogen receptor and its agonists. NEW J CHEM 2018. [DOI: 10.1039/c8nj03521k] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We present the energy profiles of estrogen receptor–agonist ligand interactions in atomic detail using a quantum biochemical approach.
Collapse
Affiliation(s)
- Aranthya H. Lima Costa
- Departamento de Biofísica e Farmacologia
- Universidade Federal do Rio Grande do Norte
- Natal-RN
- Brazil
| | - Washington S. Clemente
- Departamento de Biofísica e Farmacologia
- Universidade Federal do Rio Grande do Norte
- Natal-RN
- Brazil
| | - Katyanna S. Bezerra
- Departamento de Biofísica e Farmacologia
- Universidade Federal do Rio Grande do Norte
- Natal-RN
- Brazil
| | - José X. Lima Neto
- Departamento de Biofísica e Farmacologia
- Universidade Federal do Rio Grande do Norte
- Natal-RN
- Brazil
| | | | - Umberto L. Fulco
- Departamento de Biofísica e Farmacologia
- Universidade Federal do Rio Grande do Norte
- Natal-RN
- Brazil
| |
Collapse
|
17
|
Boroumand Moghaddam A, Moniri M, Azizi S, Abdul Rahim R, Bin Ariff A, Navaderi M, Mohamad R. Eco-Friendly Formulated Zinc Oxide Nanoparticles: Induction of Cell Cycle Arrest and Apoptosis in the MCF-7 Cancer Cell Line. Genes (Basel) 2017; 8:genes8100281. [PMID: 29053567 PMCID: PMC5664131 DOI: 10.3390/genes8100281] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 09/24/2017] [Accepted: 10/06/2017] [Indexed: 11/19/2022] Open
Abstract
Green products have strong potential in the discovery and development of unique drugs. Zinc oxide nanoparticles (ZnO NPs) have been observed to have powerful cytotoxicity against cells that cause breast cancer. The present study aims to examine the cell cycle profile, status of cell death, and pathways of apoptosis in breast cancer cells (MCF-7) treated with biosynthesized ZnO NPs. The anti-proliferative activity of ZnO NPs was determined using MTT assay. Cell cycle analysis and the mode of cell death were evaluated using a flow cytometry instrument. Quantitative real-time-PCR (qRT-PCR) was employed to investigate the expression of apoptosis in MCF-7 cells. ZnO NPs were cytotoxic to the MCF-7 cells in a dose-dependent manner. The 50% growth inhibition concentration (IC50) of ZnO NPs at 24 h was 121 µg/mL. Cell cycle analysis revealed that ZnO NPs induced sub-G1 phase (apoptosis), with values of 1.87% at 0 μg/mL (control), 71.49% at IC25, 98.91% at IC50, and 99.44% at IC75. Annexin V/propidium iodide (PI) flow cytometry analysis confirmed that ZnO NPs induce apoptosis in MCF-7 cells. The pro-apoptotic genes p53, p21, Bax, and JNK were upregulated, whereas anti-apoptotic genes Bcl-2, AKT1, and ERK1/2 were downregulated in a dose-dependent manner. The arrest and apoptosis of MCF-7 cells were induced by ZnO NPs through several signalling pathways.
Collapse
Affiliation(s)
- Amin Boroumand Moghaddam
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
- Young Research and Elite Club, Sabzevar Branch, Islamic Azad University, Sabzevar, Iran.
| | - Mona Moniri
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
- Young Research and Elite Club, Sabzevar Branch, Islamic Azad University, Sabzevar, Iran.
| | - Susan Azizi
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - Raha Abdul Rahim
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - Arbakariya Bin Ariff
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
- Bioprocessing and Biomanufacturing Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - Mohammad Navaderi
- Young Research and Elite Club, Parand Branch, Islamic Azad University, Parand, Iran.
| | - Rosfarizan Mohamad
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
- Institute of Tropical Forestry and Forest Products, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| |
Collapse
|
18
|
Chen S, Wang H, Huang YF, Li ML, Cheng JH, Hu P, Lu CH, Zhang Y, Liu N, Tzeng CM, Zhang ZM. WW domain-binding protein 2: an adaptor protein closely linked to the development of breast cancer. Mol Cancer 2017; 16:128. [PMID: 28724435 PMCID: PMC5518133 DOI: 10.1186/s12943-017-0693-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 07/10/2017] [Indexed: 01/27/2023] Open
Abstract
The WW domain is composed of 38 to 40 semi-conserved amino acids shared with structural, regulatory, and signaling proteins. WW domain-binding protein 2 (WBP2), as a binding partner of WW domain protein, interacts with several WW-domain-containing proteins, such as Yes kinase-associated protein (Yap), paired box gene 8 (Pax8), WW-domain-containing transcription regulator protein 1 (TAZ), and WW-domain-containing oxidoreductase (WWOX) through its PPxY motifs within C-terminal region, and further triggers the downstream signaling pathway in vitro and in vivo. Studies have confirmed that phosphorylated form of WBP2 can move into nuclei and activate the transcription of estrogen receptor (ER) and progesterone receptor (PR), whose expression were the indicators of breast cancer development, indicating that WBP2 may participate in the progression of breast cancer. Both overexpression of WBP2 and activation of tyrosine phosphorylation upregulate the signal cascades in the cross-regulation of the Wnt and ER signaling pathways in breast cancer. Following the binding of WBP2 to the WW domain region of TAZ which can accelerate migration, invasion and is required for the transformed phenotypes of breast cancer cells, the transformation of epithelial to mesenchymal of MCF10A is activated, suggesting that WBP2 is a key player in regulating cell migration. When WBP2 binds with WWOX, a tumor suppressor, ER transactivation and tumor growth can be suppressed. Thus, WBP2 may serve as a molecular on/off switch that controls the crosstalk between E2, WWOX, Wnt, TAZ, and other oncogenic signaling pathways. This review interprets the relationship between WBP2 and breast cancer, and provides comprehensive views about the function of WBP2 in the regulation of the pathogenesis of breast cancer and endocrine therapy in breast cancer treatment.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361005, People's Republic of China.,Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361005, People's Republic of China.,Key Laboratory for Cancer T-Cell Therapeutics and Clinical Translation (CTCTCT), Xiamen, Fujian, 361005, People's Republic of China
| | - Han Wang
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361005, People's Republic of China.,Key Laboratory for Cancer T-Cell Therapeutics and Clinical Translation (CTCTCT), Xiamen, Fujian, 361005, People's Republic of China
| | - Yu-Fan Huang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361005, People's Republic of China
| | - Ming-Li Li
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361005, People's Republic of China.,Key Laboratory for Cancer T-Cell Therapeutics and Clinical Translation (CTCTCT), Xiamen, Fujian, 361005, People's Republic of China
| | - Jiang-Hong Cheng
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361005, People's Republic of China.,Key Laboratory for Cancer T-Cell Therapeutics and Clinical Translation (CTCTCT), Xiamen, Fujian, 361005, People's Republic of China
| | - Peng Hu
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361005, People's Republic of China.,Key Laboratory for Cancer T-Cell Therapeutics and Clinical Translation (CTCTCT), Xiamen, Fujian, 361005, People's Republic of China.,INNOVA Cell Theranostics/Clinics and TRANSLA Health Group, Yangzhou, Jiangsu, People's Republic of China
| | - Chuan-Hui Lu
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361005, People's Republic of China
| | - Ya Zhang
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361005, People's Republic of China.,Key Laboratory for Cancer T-Cell Therapeutics and Clinical Translation (CTCTCT), Xiamen, Fujian, 361005, People's Republic of China
| | - Na Liu
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361005, People's Republic of China
| | - Chi-Meng Tzeng
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361005, People's Republic of China. .,Key Laboratory for Cancer T-Cell Therapeutics and Clinical Translation (CTCTCT), Xiamen, Fujian, 361005, People's Republic of China. .,INNOVA Cell Theranostics/Clinics and TRANSLA Health Group, Yangzhou, Jiangsu, People's Republic of China.
| | - Zhi-Ming Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361005, People's Republic of China. .,Teaching Hospital of Fujian Medical University, Fuzhou, Fujian, 350004, People's Republic of China.
| |
Collapse
|
19
|
Fabian CJ, Kimler BF, Zalles CM, Phillips TA, Metheny T, Petroff BK, Havighurst TC, Kim K, Bailey HH, Heckman-Stoddard BM. Clinical Trial of Acolbifene in Premenopausal Women at High Risk for Breast Cancer. Cancer Prev Res (Phila) 2015; 8:1146-55. [PMID: 26391916 DOI: 10.1158/1940-6207.capr-15-0109] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 09/07/2015] [Indexed: 12/31/2022]
Abstract
The purpose of this study was to assess the feasibility of using the selective estrogen receptor modulator (SERM) acolbifene as a breast cancer prevention agent in premenopausal women. To do so, we assessed change in proliferation in benign breast tissue sampled by random periareolar fine-needle aspiration (RPFNA) as a primary endpoint, along with changes in other risk biomarkers and objective and subjective side effects as secondary endpoints. Twenty-five women with cytologic hyperplasia ± atypia and ≥2% of breast epithelial cells staining positive for Ki-67, received 20 mg acolbifene daily for 6-8 months, and then had benign breast tissue and blood risk biomarkers reassessed. Ki-67 decreased from a median of 4.6% [interquartile range (IQR), 3.1%-8.5%] at baseline to 1.4% (IQR, 0.6%-3.5%) after acolbifene (P < 0.001; Wilcoxon signed-rank test), despite increases in bioavailable estradiol. There were also significant decreases in expression (RT-qPCR) of estrogen-inducible genes that code for pS2, ERα, and progesterone receptor (P ≤ 0.026). There was no significant change in serum IGF1, IGFBP3, IGF1:IGFBP3 ratio, or mammographic breast density. Subjective side effects were minimal with no significant increase in hot flashes, muscle cramps, arthralgias, or fatigue. Objective measures showed a clinically insignificant decrease in lumbar spine bone density (DEXA) and an increase in ovarian cysts but no change in endometrial thickness (sonography). In summary, acolbifene was associated with favorable changes in benign breast epithelial cell proliferation and estrogen-inducible gene expression but minimal side effects, suggesting a phase IIB placebo-controlled trial evaluating it further for breast cancer prevention.
Collapse
Affiliation(s)
- Carol J Fabian
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Bruce F Kimler
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas.
| | | | - Teresa A Phillips
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Trina Metheny
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Brian K Petroff
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Thomas C Havighurst
- Department of Biostatistics and Medical Informatics, University of Wisconsin Madison, Madison, Wisconsin
| | - KyungMann Kim
- Department of Biostatistics and Medical Informatics, University of Wisconsin Madison, Madison, Wisconsin
| | - Howard H Bailey
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | | |
Collapse
|
20
|
Treatment-related risk factors for arm lymphedema among long-term breast cancer survivors. J Cancer Surviv 2015; 9:422-30. [PMID: 25913877 DOI: 10.1007/s11764-014-0416-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 11/19/2014] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Treatment-related factors may increase the risk for arm lymphedema, which may occur after surgery or even many years after initial treatment for breast cancer. The association between treatment-related risk factors and development of arm lymphedema was examined for women who participated in the long-term quality of life (LTQOL) study, a 12-15-year follow-up of a breast cancer case-control study of Hispanic and non-Hispanic white women. METHODS Among 199 cases, 43 women (15 Hispanic, 28 non-Hispanic white) reported physician-diagnosed lymphedema during follow-up. Multivariable logistic regression analysis was used to calculate the odds ratios (OR) and 95% confidence intervals (CI) for the association of risk factors with lymphedema, adjusting for relevant covariates. RESULTS Tamoxifen had a non-significant, positive association with lymphedema (OR = 2.07, 95% CI 0.94-4.55, p =0.07). There were no significant associations with type of surgery, radiation, or chemotherapy. Risk was increased specifically in overweight and obese women (body mass index (BMI) > =25 kg/m(2)) treated with tamoxifen (OR = 2.62, 95% CI 0.99-6.93, p = 0.05). CONCLUSIONS This study suggests that breast cancer survivors with a BMI >25 who report the use of tamoxifen therapy may be at increased risk for arm lymphedema. IMPLICATIONS FOR CANCER SURVIVORS Larger case-control studies and clinical trials should investigate the long-term association of tamoxifen treatment with arm lymphedema especially in overweight and obese women. Lymphedema risk may be another indication to consider a weight reduction program in breast cancer survivors.
Collapse
|
21
|
Microwave-assisted synthesis, characterization and cytotoxic studies of novel estrogen receptor α ligands towards human breast cancer cells. Bioorg Med Chem Lett 2015; 25:1804-1807. [DOI: 10.1016/j.bmcl.2015.01.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/31/2014] [Accepted: 01/17/2015] [Indexed: 01/30/2023]
|
22
|
Morozkina SN, Gluzdikov IA, Drozdov AS, Selivanov SI, Kovalev RA, Filatov MV, Shavva AG. Synthesis and some biological properties of sulfamates derived from 8α-analogs of steroidal estrogens. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2015. [DOI: 10.1134/s1070428015030215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
23
|
Kim DE, Kim Y, Cho DH, Jeong SY, Kim SB, Suh N, Lee JS, Choi EK, Koh JY, Hwang JJ, Kim CS. Raloxifene induces autophagy-dependent cell death in breast cancer cells via the activation of AMP-activated protein kinase. Mol Cells 2014; 38:138-44. [PMID: 25537862 PMCID: PMC4332026 DOI: 10.14348/molcells.2015.2193] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 10/30/2014] [Accepted: 11/10/2014] [Indexed: 11/27/2022] Open
Abstract
Raloxifene is a selective estrogen receptor modulator (SERM) that binds to the estrogen receptor (ER), and exhibits potent anti-tumor and autophagy-inducing effects in breast cancer cells. However, the mechanism of raloxifene-induced cell death and autophagy is not well-established. So, we analyzed mechanism underlying death and autophagy induced by raloxifene in MCF-7 breast cancer cells. Treatment with raloxifene significantly induced death in MCF-7 cells. Raloxifene accumulated GFP-LC3 puncta and increased the level of autophagic marker proteins, such as LC3-II, BECN1, and ATG12-ATG5 conjugates, indicating activated autophagy. Raloxifene also increased autophagic flux indicators, the cleavage of GFP from GFP-LC3 and only red fluorescence-positive puncta in mRFP-GFP-LC3-expressing cells. An autophagy inhibitor, 3-methyladenine (3-MA), suppressed the level of LC3-II and blocked the formation of GFP-LC3 puncta. Moreover, siRNA targeting BECN1 markedly reversed cell death and the level of LC3-II increased by raloxifene. Besides, raloxifene-induced cell death was not related to cleavage of caspases-7, -9, and PARP. These results indicate that raloxifene activates autophagy-dependent cell death but not apoptosis. Interestingly, raloxifene decreased the level of intracellular adenosine triphosphate (ATP) and activated the AMPK/ULK1 pathway. However it was not suppressed the AKT/mTOR pathway. Addition of ATP decreased the phosphorylation of AMPK as well as the accumulation of LC3-II, finally attenuating raloxifene-induced cell death. Our current study demonstrates that raloxifene induces autophagy via the activation of AMPK by sensing decreases in ATP, and that the overactivation of autophagy promotes cell death and thereby mediates the anti-cancer effects of raloxifene in breast cancer cells.
Collapse
Affiliation(s)
- Dong Eun Kim
- Institute for Innovative Cancer Research, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Korea
| | - Yunha Kim
- Institute for Innovative Cancer Research, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Korea
| | - Dong-Hyung Cho
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin 446-701, Korea
| | - Seong-Yun Jeong
- Institute for Innovative Cancer Research, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Korea
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Korea
| | - Sung-Bae Kim
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Korea
| | - Nayoung Suh
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Korea
- Department of Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Korea
| | - Jung Shin Lee
- Institute for Innovative Cancer Research, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Korea
- Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Korea
| | - Eun Kyung Choi
- Institute for Innovative Cancer Research, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Korea
- Department of Radiation Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Korea
| | - Jae-Young Koh
- Neural Injury Research Center and Department of Neurology, Asan Medical Center, Seoul, Korea
| | - Jung Jin Hwang
- Institute for Innovative Cancer Research, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Korea
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Korea
| | - Choung-Soo Kim
- Institute for Innovative Cancer Research, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736, Korea
- Department of Urology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
24
|
Wei W, Chen ZJ, Zhang KS, Yang XL, Wu YM, Chen XH, Huang HB, Liu HL, Cai SH, Du J, Wang HS. The activation of G protein-coupled receptor 30 (GPR30) inhibits proliferation of estrogen receptor-negative breast cancer cells in vitro and in vivo. Cell Death Dis 2014; 5:e1428. [PMID: 25275589 PMCID: PMC4649509 DOI: 10.1038/cddis.2014.398] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 08/18/2014] [Accepted: 08/20/2014] [Indexed: 01/20/2023]
Abstract
There is an urgent clinical need for safe and effective treatment agents and therapy targets for estrogen receptor negative (ER−) breast cancer. G protein-coupled receptor 30 (GPR30), which mediates non-genomic signaling of estrogen to regulate cell growth, is highly expressed in ER− breast cancer cells. We here showed that activation of GPR30 by the receptor-specific agonist G-1 inhibited the growth of ER− breast cancer cells in vitro. Treatment of ER− breast cancer cells with G-1 resulted in G2/M-phase arrest, downregulation of G2-checkpoint regulator cyclin B, and induction of mitochondrial-related apoptosis. The G-1 treatment increased expression of p53 and its phosphorylation levels at Serine 15, promoted its nuclear translocation, and inhibited its ubiquitylation, which mediated the growth arrest effects on cell proliferation. Further, the G-1 induced sustained activation and nuclear translocation of ERK1/2, which was mediated by GPR30/epidermal growth factor receptor (EGFR) signals, also mediated its inhibition effects of G-1. With extensive use of siRNA-knockdown experiments and inhibitors, we found that upregulation of p21 by the cross-talk of GPR30/EGFR and p53 was also involved in G-1-induced cell growth arrest. In vivo experiments showed that G-1 treatment significantly suppressed the growth of SkBr3 xenograft tumors and increased the survival rate, associated with proliferation suppression and upregulation of p53, p21 while downregulation of cyclin B. The discovery of multiple signal pathways mediated the suppression effects of G-1 makes it a promising candidate drug and lays the foundation for future development of GPR30-based therapies for ER− breast cancer treatment.
Collapse
Affiliation(s)
- W Wei
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Z-J Chen
- Department of Pharmacy, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - K-S Zhang
- Department of Pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510120, China
| | - X-L Yang
- Key Laboratory of Tropical Disease Control (Ministry of Education), Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Institute of Human Virology, Sun Yat-sen University, Guangzhou 510655, China
| | - Y-M Wu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - X-H Chen
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - H-B Huang
- Department of Pharmacy, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - H-L Liu
- Key Laboratory of Tropical Disease Control (Ministry of Education), Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Institute of Human Virology, Sun Yat-sen University, Guangzhou 510655, China
| | - S-H Cai
- Department of Pharmacology, School of Pharmaceutical Sciences, Jinan University, Guangzhou 510632, China
| | - J Du
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - H-S Wang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
25
|
Panossian A, Hamm R, Kadioglu O, Wikman G, Efferth T. Synergy and Antagonism of Active Constituents of ADAPT-232 on Transcriptional Level of Metabolic Regulation of Isolated Neuroglial Cells. Front Neurosci 2013; 7:16. [PMID: 23430930 PMCID: PMC3576868 DOI: 10.3389/fnins.2013.00016] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 02/01/2013] [Indexed: 12/16/2022] Open
Abstract
Gene expression profiling was performed on the human neuroglial cell line T98G after treatment with adaptogen ADAPT-232 and its constituents – extracts of Eleutherococcus senticosus root, Schisandra chinensis berry, and Rhodiola rosea root as well as several constituents individually, namely, eleutheroside E, schizandrin B, salidroside, triandrin, and tyrosol. A common feature for all tested adaptogens was their effect on G-protein-coupled receptor signaling pathways, i.e., cAMP, phospholipase C (PLC), and phosphatidylinositol signal transduction pathways. Adaptogens may reduce the cAMP level in brain cells by down-regulation of adenylate cyclase gene ADC2Y and up-regulation of phosphodiesterase gene PDE4D that is essential for energy homeostasis as well as for switching from catabolic to anabolic states and vice versa. Down-regulation of cAMP by adaptogens may decrease cAMP-dependent protein kinase A activity in various cells resulting in inhibition stress-induced catabolic transformations and saving of ATP for many ATP-dependant metabolic transformations. All tested adaptogens up-regulated the PLCB1 gene, which encodes phosphoinositide-specific PLC and phosphatidylinositol 3-kinases (PI3Ks), key players for the regulation of NF-κB-mediated defense responses. Other common targets of adaptogens included genes encoding ERα estrogen receptor (2.9–22.6 fold down-regulation), cholesterol ester transfer protein (5.1–10.6 fold down-regulation), heat shock protein Hsp70 (3.0–45.0 fold up-regulation), serpin peptidase inhibitor (neuroserpin), and 5-HT3 receptor of serotonin (2.2–6.6 fold down-regulation). These findings can be reconciled with the observed beneficial effects of adaptogens in behavioral, mental, and aging-associated disorders. Combining two or more active substances in one mixture significantly changes deregulated genes profiles: synergetic interactions result in activation of genes that none of the individual substances affected, while antagonistic interactions result in suppression some genes activated by individual substances. These interactions can have an influence on transcriptional control of metabolic regulation both on the cellular level and the level of the whole organism. Merging of deregulated genes array profiles and intracellular networks is specific to the new substance with unique pharmacological characteristics. Presumably, this phenomenon could be used to eliminate undesirable effects (e.g., toxic effects) and increase the selectivity of pharmacological intervention.
Collapse
|
26
|
Hadji P, Kieback D, Tams J, Hasenburg A, Ziller M. Correlation of treatment-emergent adverse events and clinical response to endocrine therapy in early breast cancer: a retrospective analysis of the German cohort of TEAM. Ann Oncol 2012; 23:2566-2572. [DOI: 10.1093/annonc/mds055] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
27
|
Calvo E, Luu-The V, Belleau P, Martel C, Labrie F. Specific transcriptional response of four blockers of estrogen receptors on estradiol-modulated genes in the mouse mammary gland. Breast Cancer Res Treat 2012; 134:625-47. [PMID: 22678160 DOI: 10.1007/s10549-012-2104-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 05/16/2012] [Indexed: 02/01/2023]
Abstract
Novel agents for the endocrine therapy of breast cancer are needed, especially in order to take advantage of the multiple consecutive responses observed in metastatic progressing breast cancer following previous hormone therapy, thus delaying the use of cytotoxic chemotherapy with its frequent poor tolerance and serious side effects. Acolbifene (ACOL) is a novel and unique antiestrogen which represents a unique opportunity to achieve the most potent and specific blockade of estrogen action in the mammary gland and uterus while exerting estrogen-like beneficial effects in other tissues, especially the bones. To better understand the specificity of action of ACOL, we have used Affymetrix GeneChips containing 45,000 probe sets to analyze 34,000 genes to determine the specificity of this compound compared to the pure antiestrogen fulvestrant, as well as to the mixed antagonists/agonists tamoxifen and raloxifene to block the effect of estradiol (E(2)) and to induce effects of their own on the genomic profile in the mouse mammary gland. The genes modulated by E(2) were those identified in two separate experiments and validated by quantitative real-time PCR (qPCR). Three hours after the single subcutaneous injection of E(2) (0.05 μg), the simultaneous administration of ACOL, fulvestrant, tamoxifen, and raloxifene blocked by 98, 61, 43, and 92 % the number of E(2)-upregulated genes, respectively. On the other hand, 70, 10, 25, and 55 % of the genes down-regulated by E(2) were blocked by the same compounds. Of the 128 genes modulated by E(2), 49 are associated with tumorigenesis while 22 are known to be associated with breast cancer. When used alone, ACOL modulated the smallest number of genes also influenced by E(2), namely 4 %, thus possibly explaining potential utilities of this compound in breast cancer prevention and therapy.
Collapse
Affiliation(s)
- Ezequiel Calvo
- Molecular Endocrinology, Oncology and Human Genomics Research Center, Laval University and Laval University Hospital Research Center, 2705 Laurier Blvd, Quebec, QC, G1V 4G2, Canada
| | | | | | | | | |
Collapse
|
28
|
Naugler C. Estrogen receptor testing and 10-year mortality from breast cancer: A model for determining testing strategy. J Pathol Inform 2012; 3:19. [PMID: 22616031 PMCID: PMC3352613 DOI: 10.4103/2153-3539.95452] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Accepted: 04/11/2012] [Indexed: 11/22/2022] Open
Abstract
Background: The use of adjuvant tamoxifen therapy in the treatment of estrogen receptor (ER) expressing breast carcinomas represents a major advance in personalized cancer treatment. Because there is no benefit (and indeed there is increased morbidity and mortality) associated with the use of tamoxifen therapy in ER-negative breast cancer, its use is restricted to women with ER expressing cancers. However, correctly classifying cancers as ER positive or negative has been challenging given the high reported false negative test rates for ER expression in surgical specimens. In this paper I model practice recommendations using published information from clinical trials to address the question of whether there is a false negative test rate above which it is more efficacious to forgo ER testing and instead treat all patients with tamoxifen regardless of ER test results. Methods: I used data from randomized clinical trials to model two different hypothetical treatment strategies: (1) the current strategy of treating only ER positive women with tamoxifen and (2) an alternative strategy where all women are treated with tamoxifen regardless of ER test results. The variables used in the model are literature-derived survival rates of the different combinations of ER positivity and treatment with tamoxifen, varying true ER positivity rates and varying false negative ER testing rates. The outcome variable was hypothetical 10-year survival. Results: The model predicted that there will be a range of true ER rates and false negative test rates above which it would be more efficacious to treat all women with breast cancer with tamoxifen and forgo ER testing. This situation occurred with high true positive ER rates and false negative ER test rates in the range of 20-30%. Conclusions: It is hoped that this model will provide an example of the potential importance of diagnostic error on clinical outcomes and furthermore will give an example of how the effect of that error could be modeled using real-world data from clinical trials.
Collapse
Affiliation(s)
- Christopher Naugler
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, C414, Diagnostic and Scientific Centre 9, 3535 Research Road NW, Calgary AB T2L 2K8, Calgary, Canada
| |
Collapse
|
29
|
Zhan Y, Zhang Y, Liu C, Zhang J, Smith WW, Wang N, Chen Y, Zheng L, He L. A Novel Taspine Derivative, HMQ1611, Inhibits Breast Cancer Cell Growth via Estrogen Receptor α and EGF Receptor Signaling Pathways. Cancer Prev Res (Phila) 2012; 5:864-73. [DOI: 10.1158/1940-6207.capr-11-0575] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
30
|
Fourkala EO, Zaikin A, Burnell M, Gentry-Maharaj A, Ford J, Gunu R, Soromani C, Hasenbrink G, Jacobs I, Dawnay A, Widschwendter M, Lichtenberg-Fraté H, Menon U. Association of serum sex steroid receptor bioactivity and sex steroid hormones with breast cancer risk in postmenopausal women. Endocr Relat Cancer 2012; 19:137-47. [PMID: 22199143 PMCID: PMC3322660 DOI: 10.1530/erc-11-0310] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Postmenopausal women with elevated serum sex steroids have an increased risk of breast cancer. Most of this risk is believed to be exerted through binding of the sex steroids to their receptors. For the first time, we investigate the association of estrogen receptor (ER) and androgen receptor (AR) serum bioactivity (SB) in addition to hormone levels in samples from women with breast cancer collected before diagnosis. Two hundred postmenopausal women participating in the UK Collaborative Trial of Ovarian Cancer Screening who developed ER-positive breast cancer 0.6-5 years after sample donation were identified and matched to 400 controls. ER and AR bioassays were used to measure ERα, ERβ, and AR SB. Androgen and estrogen levels were measured with immunoassays. Subjects were classified according to quintiles of the respective marker among controls and the associations between SB and hormones with breast cancer risk were determined by logistic regression analysis. ERα and ERβ SB were significantly higher before diagnosis compared with controls, while estrogens showed no difference. Women had a twofold increased breast cancer risk if ERα SB (odds ratio (OR), 2.114; 95% confidence interval (CI), 1.050-4.425; P=0.040) was in the top quintile >2 years before diagnosis or estrone (OR, 2.205; 95% CI, 1.104-4.586; P=0.029) was in the top quintile <2 years before diagnosis. AR showed no significant association with breast cancer while androstenedione (OR, 3.187; 95% CI, 1.738-6.044; P=0.0003) and testosterone (OR, 2.145; 95% CI, 1.256-3.712; P=0.006) were significantly higher compared with controls and showed a strong association with an almost threefold increased breast cancer risk independent of time to diagnosis. This study provides further evidence on the association of androgens and estrogens with breast cancer. In addition, it reports that high ER but not AR SB is associated with increased breast risk >2 years before diagnosis.
Collapse
Affiliation(s)
- Evangelia-Ourania Fourkala
- Department of Gynecological Oncology, Institute for Women's Health, Gynecological Cancer Research Centre, University College London, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Khaliq W, Visvanathan K. Breast Cancer Chemoprevention: Current Approachesand Future Directions. CURRENT OBSTETRICS AND GYNECOLOGY REPORTS 2012. [DOI: 10.1007/s13669-011-0005-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
32
|
Dong X, Xiao Y, Jiang X, Wang Y. Quantitative proteomic analysis revealed lovastatin-induced perturbation of cellular pathways in HL-60 cells. J Proteome Res 2011; 10:5463-71. [PMID: 21967149 DOI: 10.1021/pr200718p] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Lovastatin, a member of the statin family of drugs, is widely prescribed for treating hypercholesterolemia. The statin family of drugs, however, also shows promise for cancer treatment and prevention. Although lovastatin is known to be an inhibitor for HMG-CoA reductase, the precise mechanisms underlying the drug's antiproliferative activity remain unclearly defined. Here we utilized mass spectrometry, in conjunction with stable isotope labeling by amino acids in cell culture (SILAC), to analyze the perturbation of protein expression in HL-60 cells treated with lovastatin. We were able to quantify ∼3200 proteins with both forward and reverse SILAC labeling experiments, among which ∼120 exhibited significant alterations in expression levels upon lovastatin treatment. Apart from confirming the expected inhibition of the cholesterol biosynthesis pathway, our quantitative proteomic results revealed that lovastatin perturbed the estrogen receptor signaling pathway, which was manifested by the diminished expression of estrogen receptor α, steroid receptor RNA activator 1, and other related proteins. Lovastatin also altered glutamate metabolism through down-regulation of glutamine synthetase and γ-glutamylcysteine synthetase. Moreover, lovastatin treatment led to a marked down-regulation of carbonate dehydratase II (a.k.a. carbonic anhydrase II) and perturbed the protein ubiquitination pathway. Together, the results from the present study underscored several new cellular pathways perturbed by lovastatin.
Collapse
Affiliation(s)
- Xiaoli Dong
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | | | | | | |
Collapse
|
33
|
Hormone replacement therapy and women with premature menopause--a cancer survivorship issue. Eur J Cancer 2011; 47:1623-32. [PMID: 21561765 DOI: 10.1016/j.ejca.2011.04.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 07/02/2011] [Accepted: 04/04/2011] [Indexed: 11/21/2022]
Abstract
The importance of addressing survivorship issues has been emphasised in recent years. As cancer therapies improve there is a growing population of cancer survivors, which includes many women with premature menopause. Women who are premenopausal at the time of their cancer diagnosis may have specific survivorship issues to be addressed, including infertility, early menopause and sexual dysfunction. These factors can continue have a significant impact on the quality of life of these patients at long term follow up. Data for this Review were identified by searches of MEDLINE, PubMed, and references from relevant articles using the search terms 'HRT', 'women/female cancer/tumour', 'menopause' and 'survivorship'. Abstracts and reports from meetings were excluded. Only papers published in English between 1980 and 2010 were included. The aims of this review are to: • Address the hormonal factors which impact on cancer survivorship for premenopausal women • Review the debate for the role of hormone replacement therapy (HRT) in cancer survivors • Provide information for physicians and patients regarding the management of hormonally driven survivorship issues (for different tumour types), based on current evidence The recommendations for practice are that HRT may be offered for the alleviation of vasomotor symptoms in cancer survivors who undergo premature menopause up to the age of natural menopause (51 years in the UK). HRT (including vaginal oestrogen preparations) is contraindicated in survivors of oestrogen receptor positive breast cancer and low grade endometrial leiomyosarcoma, where non-HRT alternatives should be considered to alleviate symptoms.
Collapse
|
34
|
Lappano R, Recchia AG, De Francesco EM, Angelone T, Cerra MC, Picard D, Maggiolini M. The cholesterol metabolite 25-hydroxycholesterol activates estrogen receptor α-mediated signaling in cancer cells and in cardiomyocytes. PLoS One 2011; 6:e16631. [PMID: 21304949 PMCID: PMC3031608 DOI: 10.1371/journal.pone.0016631] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 12/27/2010] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The hydroxylated derivatives of cholesterol, such as the oxysterols, play important roles in lipid metabolism. In particular, 25-hydroxycholesterol (25 HC) has been implicated in a variety of metabolic events including cholesterol homeostasis and atherosclerosis. 25 HC is detectable in human plasma after ingestion of a meal rich in oxysterols and following a dietary cholesterol challenge. In addition, the levels of oxysterols, including 25 HC, have been found to be elevated in hypercholesterolemic serum. METHODOLOGY/PRINCIPAL FINDINGS Here, we demonstrate that the estrogen receptor (ER) α mediates gene expression changes and growth responses induced by 25 HC in breast and ovarian cancer cells. Moreover, 25 HC exhibits the ERα-dependent ability like 17 β-estradiol (E2) to inhibit the up-regulation of HIF-1α and connective tissue growth factor by hypoxic conditions in cardiomyocytes and rat heart preparations and to prevent the hypoxia-induced apoptosis. CONCLUSIONS/SIGNIFICANCE The estrogen action exerted by 25 HC may be considered as an additional factor involved in the progression of breast and ovarian tumors. Moreover, the estrogen-like activity of 25 HC elicited in the cardiovascular system may play a role against hypoxic environments.
Collapse
Affiliation(s)
- Rosamaria Lappano
- Department of Pharmaco-Biology, University of Calabria, Rende, Italy
| | | | | | - Tommaso Angelone
- Department of Cell Biology, University of Calabria, Rende, Italy
| | | | - Didier Picard
- Département de Biologie Cellulaire, Université de Genève, Genève, Switzerland
| | - Marcello Maggiolini
- Department of Pharmaco-Biology, University of Calabria, Rende, Italy
- * E-mail:
| |
Collapse
|
35
|
Kieback D, Harbeck N, Bauer W, Hadji P, Weyer G, Menschik T, Hasenburg A. Endometrial effects of exemestane compared to tamoxifen within the Tamoxifen Exemestane Adjuvant Multicenter (TEAM) trial: Results of a prospective gynecological ultrasound substudy. Gynecol Oncol 2010; 119:500-5. [DOI: 10.1016/j.ygyno.2010.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Revised: 08/05/2010] [Accepted: 08/06/2010] [Indexed: 10/19/2022]
|
36
|
Kretzer NM, Cherian MT, Mao C, Aninye IO, Reynolds PD, Schiff R, Hergenrother PJ, Nordeen SK, Wilson EM, Shapiro DJ. A noncompetitive small molecule inhibitor of estrogen-regulated gene expression and breast cancer cell growth that enhances proteasome-dependent degradation of estrogen receptor {alpha}. J Biol Chem 2010; 285:41863-73. [PMID: 21041310 DOI: 10.1074/jbc.m110.183723] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The mechanisms responsible for 17β-estradiol (E(2))-stimulated breast cancer growth and development of resistance to tamoxifen and other estrogen receptor α (ERα) antagonists are not fully understood. We describe a new tool for dissecting ERα action in breast cancer, p-fluoro-4-(1,2,3,6,-tetrahydro-1,3-dimethyl-2-oxo-6-thionpurin-8-ylthio) (TPSF), a potent small-molecule inhibitor of estrogen receptor α that does not compete with estrogen for binding to ERα. TPSF noncompetitively inhibits estrogen-dependent ERα-mediated gene expression with little inhibition of transcriptional activity by NF-κB or the androgen or glucocorticoid receptor. TPSF inhibits E(2)-ERα-mediated induction of the proteinase inhibitor 9 gene, which is activated by ERα binding to estrogen response element DNA, and the cyclin D1 gene, which is induced by tethering ERα to other DNA-bound proteins. TPSF inhibits anchorage-dependent and anchorage-independent E(2)-ERα-stimulated growth of MCF-7 cells but does not inhibit growth of ER-negative MDA-MB-231 breast cancer cells. TPSF also inhibits ERα-dependent growth in three cellular models for tamoxifen resistance; that is, 4-hydroxytamoxifen-stimulated MCF7ERαHA cells that overexpress ERα, fully tamoxifen-resistant BT474 cells that have amplified HER-2 and AIB1, and partially tamoxifen-resistant ZR-75 cells. TPSF reduces ERα protein levels in MCF-7 cells and several other cell lines without altering ERα mRNA levels. The proteasome inhibitor MG132 abolished down-regulation of ERα by TPSF. Thus, TPSF affects receptor levels at least in part due to its ability to enhance proteasome-dependent degradation of ERα. TPSF represents a novel class of ER inhibitor with significant clinical potential.
Collapse
Affiliation(s)
- Nicole M Kretzer
- Department of Biochemistry, University of Illinois, Urbana, Illinois 61801-3602, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Amin ARMR, Wang D, Zhang H, Peng S, Shin HJC, Brandes JC, Tighiouart M, Khuri FR, Chen ZG, Shin DM. Enhanced anti-tumor activity by the combination of the natural compounds (-)-epigallocatechin-3-gallate and luteolin: potential role of p53. J Biol Chem 2010; 285:34557-65. [PMID: 20826787 DOI: 10.1074/jbc.m110.141135] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Natural dietary agents have drawn a great deal of attention toward cancer prevention because of their wide safety margin. However, single agent intervention has failed to bring the expected outcome in clinical trials; therefore, combinations of chemopreventive agents are gaining increasingly popularity. In the present study, we investigated a combinatorial approach using two natural dietary polyphenols, luteolin and EGCG, and found that their combination at low doses (at which single agents induce minimal apoptosis) synergistically increased apoptosis (3-5-fold more than the additive level of apoptosis) in both head and neck and lung cancer cell lines. This combination also significantly inhibited growth of xenografted tumors in nude mice. The in vivo findings also were supported by significant inhibition of Ki-67 expression and increase in TUNEL-positive cells in xenografted tissues. Mechanistic studies revealed that the combination induced mitochondria-dependent apoptosis in some cell lines and mitochondria-independent apoptosis in others. Moreover, we found more efficient stabilization and ATM-dependent Ser(15) phosphorylation of p53 due to DNA damage by the combination, and ablation of p53 using shRNA strongly inhibited apoptosis as evidenced by decreased poly(ADP-ribose) polymerase and caspase-3 cleavage. In addition, we observed mitochondrial translocation of p53 after treatment with luteolin or the combination of EGCG and luteolin. Taken together, our results for the first time suggest that the combination of luteolin and EGCG has synergistic/additive growth inhibitory effects and provides an important rationale for future chemoprevention trials of head and neck and lung cancers.
Collapse
Affiliation(s)
- A R M Ruhul Amin
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Masri S, Phung S, Wang X, Chen S. Molecular characterization of aromatase inhibitor-resistant, tamoxifen-resistant and LTEDaro cell lines. J Steroid Biochem Mol Biol 2010; 118:277-82. [PMID: 19897035 PMCID: PMC2836255 DOI: 10.1016/j.jsbmb.2009.10.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Accepted: 10/27/2009] [Indexed: 01/07/2023]
Abstract
To determine potential genes involved in mediating resistance to aromatase inhibitors (AIs), a microarray study was performed using MCF-7aro (aromatase overexpressing) cells that are resistant to letrozole (T+LET R), anastrozole (T+ANA R) and exemestane (T+EXE R), as well as LTEDaro and tamoxifen-resistant (T+TAM R) lines for comparison. Based on hierarchical clustering, estrogen-responsive genes were found to be differentially expressed in AI-resistant lines versus LTEDaro and T+TAM R. Additional genome-wide analysis showed that gene expression profiles of the non-steroidal AI-resistant lines were most closely correlated and that T+EXE R lines exhibit differing profiles. Also, LTEDaro and T+TAM R lines are inherently different from expression profiles of AI-resistant lines. Further characterization of these resistant lines revealed that T+LET R, T+ANA R and LTEDaro cells contain a constitutively active estrogen receptor alpha (ERalpha) that does not require the ligand estrogen for activation. Ligand-independent activation of ERalpha does not activate identical estrogen-responsive gene profiles in AI-resistant lines as in LTEDaro lines, thereby establishing differing mechanisms of resistance. This ligand-independent activation of ER was not observed in the parental cell lines MCF-7aro, T+EXE R or T+TAM R cells. Based on the steroidal structure of EXE, our laboratory has shown that this AI has weak estrogen-like properties, and that EXE resistance involves an ER-dependent crosstalk with EGFR growth factor signaling. Recent studies in our laboratory pertaining to pre-clinical models of AI treatment revealed that intermittent use of EXE delays the onset of acquired resistance in comparison to continuous treatment. Specific molecular mechanisms involved in intermittent use of EXE are currently being explored, based on microarray gene expression profiling. Lastly, our laboratory has initiated a study of microRNAs and their potential role in regulating target genes involved in AI-resistance. Overall, we propose a model of acquired resistance that progresses from hormone-dependence (T+TAM R and T+EXE R) to hormone-independence (T+LET R and T+ANA R), eventually resulting in hormone-independence that does not rely on conventional ER signaling (LTEDaro).
Collapse
Affiliation(s)
- Selma Masri
- Division of Tumor Cell Biology, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | | | | | | |
Collapse
|
39
|
Rashid F, Khan RN, Iftikhar SY. Probing the link between oestrogen receptors and oesophageal cancer. World J Surg Oncol 2010; 8:9. [PMID: 20146809 PMCID: PMC2831901 DOI: 10.1186/1477-7819-8-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 02/10/2010] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Human oesophageal carcinoma is considered to be one of the most aggressive malignancies and has a very poor prognosis. The incidence of oesophageal cancer shows a gender bias and is higher in males compared with females, the ratio between males and females varying from 3:1 to 7:1. This sex ratio is not entirely attributable to differences in the prevalence of known risk factors between the sexes. The potential role of oestrogen receptors (ER) in oesophageal cancer has been debated for several years but the significance of the receptors in this cancer remains unknown. Most of the work has been based on immunohistochemistry and has not been validated with other available techniques. The inconsistencies in the published literature on the link between ER expression and oesophageal cancer warrant a thorough evaluation of the potential role of ERs in this malignancy. Even the expression of the two ER isoforms, ERalpha and ERbeta, and its implications for outcome of treatments in histological subtypes of oesophageal tumours is ill defined. The aim of this article is to provide updated information from the available literature on the current status of ER expression in oesophageal cancer and to discuss its potential therapeutic role. METHODS AND RESULTS We performed a comprehensive literature search and analysed the results regarding ER expression in oesophageal tumours with special emphasis on expression of different oestrogen receptors and the role of sex hormones in oesophageal cancer. This article also focuses on the significance of the two main ER subtypes and mechanisms underlying the presumed male predominance of this disease. CONCLUSION We postulate that differential oestrogen receptor status may be considered a biomarker of poor clinical outcome based on tissue dedifferentiation or advanced stage of the disease. Further, if we can establish the importance of oestrogen and its receptors in the context of oesophageal cancer, then this may lead to a new future direction in the management of this malignancy.
Collapse
Affiliation(s)
- Farhan Rashid
- Department of Upper GI Surgery, Royal Derby Hospital, Uttoxeter Road, Derby DE22 3NE, UK.
| | | | | |
Collapse
|
40
|
Sarkar S, Mandal M. Growth factor receptors and apoptosis regulators: signaling pathways, prognosis, chemosensitivity and treatment outcomes of breast cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2009; 3:47-60. [PMID: 21556249 PMCID: PMC3086304 DOI: 10.4137/bcbcr.s2492] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Biomarkers of breast cancer are necessary for prognosis and prediction to chemotherapy. Prognostic biomarkers provide information regarding outcome irrespective of therapy, while predictive biomarkers provide information regarding response to therapy. Candidate prognostic biomarkers for breast cancers are growth factor receptors, steroid receptors, Ki-67, cyclins, urokinase plasminogen activator, p53, p21, pro- and anti-apoptotic factors, BRCA1 and BRCA2. But currently, the predictive markers are Estrogen and Progesterone receptors responding to endocrine therapy, and HER-2 responding to herceptin. But there are numerous breast cancer cases, where tamoxifen is ineffective even after estrogen receptor positivity. This lead to search of new prognostic and predictive markers and the number of potential markers is constantly increasing due to proteomics and genomics studies. However, most biomarkers individually have poor sensitivity or specificity, or other clinical value. It can be resolved by studying various biomarkers simultaneously, which will help in better prognosis and increasing sensitivity for chemotherapeutic agents. This review is focusing on growth factor receptors, apoptosis markers, signaling cascades, and their correlation with other associated biomarkers in breast cancers. As our knowledge regarding molecular biomarkers for breast cancer increases, prognostic indices will be developed that combine the predictive power of individual molecular biomarkers with specific clinical and pathologic factors. Rigorous comparison of these existing as well as emerging markers with current treatment selection is likely to see an escalation in an era of personalized medicines to ensure the breast cancer patients receive optimal treatment. This will also solve the treatment modalities and complications related to chemotherapeutic regimens.
Collapse
Affiliation(s)
- Siddik Sarkar
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur-721302, India
| | | |
Collapse
|
41
|
Fabian CJ. Surrogate response biomarkers in prevention research: do they point the way or lead us astray? J Clin Oncol 2009; 27:3734-6. [PMID: 19597020 DOI: 10.1200/jco.2009.22.9211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
42
|
William WN, Heymach JV, Kim ES, Lippman SM. Molecular targets for cancer chemoprevention. Nat Rev Drug Discov 2009; 8:213-25. [DOI: 10.1038/nrd2663] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
43
|
Planas-Silva MD, Rutherford TM, Stone MC. Prevention of age-related spontaneous mammary tumors in outbred rats by late ovariectomy. ACTA ACUST UNITED AC 2008; 32:65-71. [PMID: 18407436 DOI: 10.1016/j.cdp.2008.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2008] [Indexed: 11/29/2022]
Abstract
BACKGROUND Breast cancer prevention trials have shown that the antiestrogen tamoxifen inhibits development of estrogen receptor (ER)-positive tumors. In Sprague-Dawley rats, removal of ovarian function in young animals can reduce the incidence of spontaneous age-dependent mammary tumors. However, it is not known whether removal of ovaries late in life, before middle age onset, can still prevent mammary tumor development. METHODS In this study we used Hsd:Sprague-Dawley SD (Hsd) rats to determine the effect of late ovariectomy on mammary tumor development. Intact, sham-ovariectomized and ovariectomized rats were followed until 110 weeks of age, or over their life span. In some experiments, palpable tumors were surgically removed upon presentation. RESULTS Removal of ovaries before middle age onset ( approximately 5-7 months) inhibited development of spontaneous mammary tumors by 95%. Only one mammary tumor was observed in 19 late ovariectomized animals while 47 total tumors developed in 42 non-ovariectomized animals. Tumor incidence was reduced from 73.8 to 5.3% (relative risk=0.05, 95% CI=0.0072-0.354). The frequency of mammary carcinomas in non-ovariectomized virgin female rats was one in eight rats. Spontaneous rat carcinomas expressed ER and other biomarkers, such as cyclin D1. When palpable tumors were removed by surgical excision, tumor multiplicity increased from 0.76 to 1.61 tumors per rat. Surprisingly, ovariectomy increased the 110-week survival rate and maximum life span of Hsd rats. CONCLUSION Late ovariectomy prevents spontaneous mammary tumor development in Hsd rats. This animal model may be useful for evaluating novel interventions in breast cancer prevention.
Collapse
Affiliation(s)
- Maricarmen D Planas-Silva
- Department of Pharmacology, Penn State College of Medicine, MCH078, 500 University Drive, Hershey, PA 17033, USA.
| | | | | |
Collapse
|
44
|
Minutolo F, Bellini R, Bertini S, Carboni I, Lapucci A, Pistolesi L, Prota G, Rapposelli S, Solati F, Tuccinardi T, Martinelli A, Stossi F, Carlson KE, Katzenellenbogen BS, Katzenellenbogen JA, Macchia M. Monoaryl-Substituted Salicylaldoximes as Ligands for Estrogen Receptor β. J Med Chem 2008; 51:1344-51. [DOI: 10.1021/jm701396g] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Filippo Minutolo
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - Rosalba Bellini
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - Simone Bertini
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - Isabella Carboni
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - Annalina Lapucci
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - Letizia Pistolesi
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - Giovanni Prota
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - Simona Rapposelli
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - Francesca Solati
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - Tiziano Tuccinardi
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - Adriano Martinelli
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - Fabio Stossi
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - Kathryn E. Carlson
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - Benita S. Katzenellenbogen
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - John A. Katzenellenbogen
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| | - Marco Macchia
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy, and Department of Molecular and Integrative Physiology and Department of Chemistry, University of Illinois, 600 S. Goodwin Avenue, Urbana, Illinois 61801
| |
Collapse
|
45
|
Abstract
Colorectal cancer is the third most prevalent cancer in the world. If detected at an early stage, treatment often might lead to cure. As prevention is better than cure, epidemiological studies reveal that having a healthy diet often protects from promoting/ developing cancer. An important consideration in evaluating new drugs and devices is determining whether a product can effectively treat a targeted disease. There are quite a number of biomarkers making their way into clinical trials and few are awaiting the preclinical efficacy and safety results to enter into clinical trials. Researchers are facing challenges in modifying trial design and defining the right control population, validating biomarker assays from the biological and analytical perspective and using biomarker data as a guideline for decision making. In spite of following all guidelines, the results are disappointing from many of the large clinical trials. To avoid these disappointments, selection of biomarkers and its target drug needs to be evaluated in appropriate animal models for its toxicities and efficacies. The focus of this review is on the few of the potential molecular targets and their biomarkers in colorectal cancers. Strengths and limitations of biomarkers/surrogate endpoints are also discussed. Various pathways involved in tumor cells and the specific agents to target the altered molecular biomarker in biomolecular pathway are elucidated. Importance of emerging new platforms siRNAs and miRNAs technology for colorectal cancer therapeutics is reviewed.
Collapse
Affiliation(s)
- Naveena B Janakiram
- Department of Medicine, Hem-Onc Section, OU Cancer Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | |
Collapse
|
46
|
|
47
|
Abstract
The third-generation aromatase inhibitors (AIs) anastrozole, exemestane and letrozole have largely replaced tamoxifen as the preferred treatment for hormone receptor - positive breast cancer in postmenopausal women. Approximately 185,000 new cases of invasive breast cancer are diagnosed yearly, and at least half of these women are both postmenopausal and eligible for adjuvant therapy with AIs. In addition, AIs are currently being tested as primary prevention therapy in large randomised trials involving tens of thousands of women at increased risk for breast cancer. Given the volume of use, internists will increasingly see postmenopausal women who are taking or considering treatment with AIs. Physicians need to be able to: (i) briefly discuss the pros and cons of using a selective estrogen receptor modulator such as tamoxifen or raloxifene vs. an AI for risk reduction and (ii) recognise and manage AI-associated adverse events. The primary purpose of this review is to help internists with these two tasks.
Collapse
Affiliation(s)
- C J Fabian
- Breast Cancer Prevention Center, Division of Clinical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160-7418, USA.
| |
Collapse
|
48
|
Goldenberg VK, Seewaldt VL, Scott V, Bean GR, Broadwater G, Fabian C, Kimler B, Zalles C, Lipkus IM. Atypia in random periareolar fine-needle aspiration affects the decision of women at high risk to take tamoxifen for breast cancer chemoprevention. Cancer Epidemiol Biomarkers Prev 2007; 16:1032-4. [PMID: 17507634 DOI: 10.1158/1055-9965.epi-06-0910] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Random periareolar fine-needle aspiration (RPFNA) is a research procedure designed to (a) evaluate short-term breast cancer risk in women at high risk for developing breast cancer, and (b) track response to chemoprevention. Of import, cellular atypia in breast RPFNA is prospectively associated with a 5.6-fold increase in breast cancer risk in women at high risk. Among 99 women attending a clinic for high-risk breast cancer, we explored the effects of RPFNA cytology results on decision making pertaining to the use of tamoxifen for breast cancer chemoprevention. No patient with nonproliferative or hyperplastic cytology subsequently elected to take tamoxifen. Only 7% of subjects with borderline atypia elected to take tamoxifen. In contrast, 50% with atypia elected to take tamoxifen. These results suggest that the provision of a biomarker of short-term risk can affect the motivation to take tamoxifen for chemoprevention. This conclusion is informative given that tamoxifen, due to its side effects, is often underused by women at high risk of developing breast cancer. Further research is needed to determine the mechanisms through which RPFNA results affect the decision to use tamoxifen, or any other breast cancer chemopreventive agent.
Collapse
MESH Headings
- Adult
- Anticarcinogenic Agents/therapeutic use
- Biopsy, Fine-Needle/methods
- Breast/cytology
- Breast/pathology
- Breast Neoplasms/diagnosis
- Breast Neoplasms/pathology
- Breast Neoplasms/prevention & control
- Carcinoma, Ductal, Breast/diagnosis
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/prevention & control
- Carcinoma, Lobular/diagnosis
- Carcinoma, Lobular/pathology
- Carcinoma, Lobular/prevention & control
- Cohort Studies
- Decision Making
- Female
- Humans
- Middle Aged
- Nipples
- Risk Factors
- Selective Estrogen Receptor Modulators/therapeutic use
- Tamoxifen/therapeutic use
Collapse
|
49
|
Abstract
Epidemiologic models used for cancer risk prediction, such as the Gail model, are validated for populations undergoing regular screening but often have suboptimal individual predictive accuracy. Risk biomarkers may be employed to improve predictive accuracy based on the Gail or other epidemiologic models and, to the extent that they are reversible, may be used to assess response in phase I-II prevention trials. Risk biomarkers used as intermediate response endpoints include high mammographic breast density, intra-epithelial neoplasia, and cytomorphology with associated molecular markers such as Ki-67. At the present time these biomarkers may not be used to predict or monitor individual response to standard prevention interventions but are used in early phase clinical trials as preliminary indicators of efficacy.
Collapse
Affiliation(s)
- Carol J Fabian
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA.
| | | |
Collapse
|
50
|
Weitzel JN, Buys SS, Sherman WH, Daniels A, Ursin G, Daniels JR, MacDonald DJ, Blazer KR, Pike MC, Spicer DV. Reduced mammographic density with use of a gonadotropin-releasing hormone agonist-based chemoprevention regimen in BRCA1 carriers. Clin Cancer Res 2007; 13:654-8. [PMID: 17255289 DOI: 10.1158/1078-0432.ccr-06-1902] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Women with a BRCA1 mutation (BRCA1(mut)) need risk reduction options beyond mastectomy and oophorectomy. We evaluated the efficacy, safety, and tolerability of hormonal chemoprevention with a gonadotropin-releasing hormone agonist (GnRHA) with low-dose add-back steroids in BRCA1(mut) carriers. EXPERIMENTAL DESIGN The 12-month open label clinical trial used the GnRHA deslorelin, ultra-low-dose estradiol (E(2)), and replacement testosterone, administered via daily intranasal spray in premenopausal women with a BRCA1(mut), and intermittent oral medroxyprogesterone acetate. The end points included mammographic percent density, bone mineral density, endometrial hyperplasia, symptom inventory, and quality of life (Medical Outcomes SF-36 survey). RESULTS Six of eight BRCA1(mut) women (mean age, 30.3 years; range, 25-36 years) completed the study. Mammographic percent density was significantly reduced at 12 months (median absolute mammographic percent density decrease, 8.3%; P = 0.043), representing a 29.2% median reduction in mammographic percent density. Bone mineral density remained within reference limits for all participants; there were no cases of atypical endometrial hyperplasia and menses resumed within a median of 67 days (range, 35-110 days) after last drug treatment day. The treatment was well tolerated; hypoestrogenic side effects were minimal and transient; and there were no significant changes in quality of life. CONCLUSIONS The GnRHA deslorelin, with low-dose add-back steroids, was well tolerated and significantly decreased mammographic percent density in BRCA1(mut) carriers. This regimen may reduce breast cancer risk and improve the usefulness of mammographic surveillance by reducing density. This is the first demonstration, to our knowledge, of a direct reduction of mammographic densities in young BRCA1(mut) carriers.
Collapse
Affiliation(s)
- Jeffrey N Weitzel
- Department of Clinical Cancer Genetics, City of Hope Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|