251
|
Wang M, Xia F, Wei Y, Wei X. Molecular mechanisms and clinical management of cancer bone metastasis. Bone Res 2020; 8:30. [PMID: 32793401 PMCID: PMC7391760 DOI: 10.1038/s41413-020-00105-1] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/03/2019] [Accepted: 10/23/2019] [Indexed: 02/05/2023] Open
Abstract
As one of the most common metastatic sites of malignancies, bone has a unique microenvironment that allows metastatic tumor cells to grow and flourish. The fenestrated capillaries in the bone, bone matrix, and bone cells, including osteoblasts and osteoclasts, together maintain the homeostasis of the bone microenvironment. In contrast, tumor-derived factors act on bone components, leading to subsequent bone resorption or excessive bone formation. The various pathways involved also provide multiple targets for therapeutic strategies against bone metastases. In this review, we summarize the current understanding of the mechanism of bone metastases. Based on the general process of bone metastases, we specifically highlight the complex crosstalk between tumor cells and the bone microenvironment and the current management of cancer bone metastases.
Collapse
Affiliation(s)
- Manni Wang
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| | - Fan Xia
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan P.R. China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| |
Collapse
|
252
|
Hahm ER, Kim SH, Singh KB, Singh K, Singh SV. A Comprehensive Review and Perspective on Anticancer Mechanisms of Withaferin A in Breast Cancer. Cancer Prev Res (Phila) 2020; 13:721-734. [PMID: 32727824 DOI: 10.1158/1940-6207.capr-20-0259] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/23/2020] [Accepted: 07/22/2020] [Indexed: 01/07/2023]
Abstract
Withaferin A (hereafter abbreviated as WA) is a promising anticancer steroidal lactone abundant in a medicinal plant (Withania somnifera) native to Asia. The root/leaf extract of Withania somnifera, which belongs to the Solanaceae family, continues to be included in the Ayurvedic medicine formulations of alternative medicine practice. Numerous chemicals are detectable in the root/leaf extract of Withania somnifera [e.g., withanolides (WA, withanone, withanolide A, etc.), alkaloids, sitoindosides, etc.], but the anticancer effect of this medicinal plant is largely attributed to WA. Anticancer effect of WA was initially reported in the early 70s in the Ehrlich ascites tumor cell model in vitro Since then, numerous preclinical studies have been performed using cellular and animal models of different cancers including breast cancer to determine cancer therapeutic and chemopreventive effects of WA. Chemoprevention, a word first introduced by Dr. Michael B. Sporn, was intended to impede, arrest, or reverse carcinogenesis at its earliest stages with pharmacologic agents. This review succinctly summarizes the published findings on anticancer pharmacology of WA in breast cancer focusing on pharmacokinetic behavior, in vivo efficacy data in preclinical models in a therapeutic and chemoprevention settings, and its known effects on cancer-relevant cellular processes (e.g., growth arrest, apoptosis induction, autophagy, metabolic adaptation, immune function, etc.) and molecular targets (e.g., suppression of oncogenes such as estrogen receptor-α, STAT3, etc.). Potential gaps in knowledge as well as future research directions essential for clinical development of WA for chemoprevention and/or treatment of breast cancer are also discussed.
Collapse
Affiliation(s)
- Eun-Ryeong Hahm
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Su-Hyeong Kim
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Krishna B Singh
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kamayani Singh
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shivendra V Singh
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania. .,UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
253
|
Fatigante M, Heritage J, Alby F, Zucchermaglio C. Presenting treatment options in breast cancer consultations: Advice and consent in Italian medical care. Soc Sci Med 2020; 266:113175. [PMID: 32987310 DOI: 10.1016/j.socscimed.2020.113175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/11/2020] [Accepted: 06/25/2020] [Indexed: 01/05/2023]
Abstract
Variety, complexity and uncertainty in the therapy outcomes of cancer illness make the treatment recommendation (TR) in oncology a "monumentally difficult task". Previous studies have distinguished unilateral and bilateral formats of treatment recommendations, accordingly to whether, or to what extent, the patient's perspective is included in the consideration of therapeutic options. Others have also shown how the oncologists' presentation of therapeutic options varied accordingly to the severity of the diagnosis and the availability of alternatives. Yet, no study has systemically dentified and compared components of treatment recommendation in oncology on a common set of patients and clinicians. This paper analyzes how different options in breast cancer treatments - radiotherapy, hormone therapy and chemotherapy - are presented and discussed in a set of 12 first post-surgical breast cancer visits carried out by two oncologists of high experience and seniority in two Italian hospitals. Treatment recommendation sequences involving these three option types were analyzed using the methods of conversation analysis. They were also coded for the mention of side effects and treatment burden, and for whether consent to the recommendation was invited, or expressed by the patient. Results show that radiotherapy is presented as presupposed as an extension of surgery and is not further discussed, and hormone therapy is delivered as good news and as not implying any health or lifestyle burdens. Treatment burdens were raised in the much more extensive discussions of chemotherapy, which were also accompanied by a higher chance that the patient was asked for consent to therapy. Implications are drawn as regards the extent to which clinical practice meets theory in communication protocols available in oncology, and how to consider the doctor-patient partnership and the concept of shared decision-making in such an encounter.
Collapse
Affiliation(s)
- Marilena Fatigante
- Department of Social and Developmental Psychology, University Sapienza of Rome, Rome, Italy.
| | - John Heritage
- Department of Sociology, University of California, Los Angeles, USA
| | - Francesca Alby
- Department of Social and Developmental Psychology, University Sapienza of Rome, Rome, Italy
| | - Cristina Zucchermaglio
- Department of Social and Developmental Psychology, University Sapienza of Rome, Rome, Italy
| |
Collapse
|
254
|
Ignatov A, Ortmann O. Endocrine Risk Factors of Endometrial Cancer: Polycystic Ovary Syndrome, Oral Contraceptives, Infertility, Tamoxifen. Cancers (Basel) 2020; 12:E1766. [PMID: 32630728 PMCID: PMC7408229 DOI: 10.3390/cancers12071766] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022] Open
Abstract
Endometrial cancer is the most common gynecologic cancer and is predominantly endocrine-related. The role of unopposed estrogen in the development of endometrial cancer has been investigated in numerous studies. Different reproductive factors such as younger age at menarche, late age at menopause, infertility, nulliparity, age of birth of the first child, and long-term use of unopposed estrogens during hormone replacement therapy have been associated with an increased risk of endometrial cancer. In contrast, there is a growing body of evidence for a protective role of oral contraceptives. Most of the published data on the association between infertility and polycystic ovary syndrome are inconclusive, whereas the effect of tamoxifen on the risk of endometrial cancer has been well established. With this review, we aim to summarize the evidence on the association between infertility, polycystic ovary syndrome, oral contraceptives, and tamoxifen and the development of endometrial cancer.
Collapse
Affiliation(s)
- Atanas Ignatov
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Landshuter Str. 65, 93053 Regensburg, Germany;
| | | |
Collapse
|
255
|
Chlebowski RT, Aragaki AK, Anderson GL, Prentice RL. Forty-year trends in menopausal hormone therapy use and breast cancer incidence among postmenopausal black and white women. Cancer 2020; 126:2956-2964. [PMID: 32212335 PMCID: PMC11995946 DOI: 10.1002/cncr.32846] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 09/22/2019] [Accepted: 10/21/2019] [Indexed: 01/22/2023]
Abstract
After reports from the Women's Health Initiative randomized trial evaluating estrogen plus progestin, there was a sudden, substantial, and sustained decrease in all categories of menopausal hormone therapy, and the first reduction in age-adjusted breast cancer incidence in more than 20 years was seen in 2003-2004 among US women 50 years of age or older. Subsequent trends in breast cancer incidence have been described, but most reports have not focused on the postmenopausal age group or fully engaged the potential influence of reduced hormone therapy on breast cancer incidence trends by race/ethnicity. To address this gap, this commentary examines trends for annual age-adjusted breast cancer incidence over a 40-year period from 1975 to 2015 for white and black women on the basis of findings from the Surveillance, Epidemiology, and End Results 9 registries. Overall, the sharp decline in breast cancer incidence seen in 2003-2004 was followed in the subsequent decade by a continued low breast cancer incidence plateau in white women that has largely persisted. In contrast, a new discordance between breast cancer incidence trends in black and white women has emerged. In the 2005-2015 decade, a sustained increase in breast cancer incidence in black women has resulted in annual incidence rates comparable, for the first time, to those in white women. This commentary explores the hypothesis that the over-decade-long and discordant changes in breast cancer incidence rates in postmenopausal black and white women are, to a large extent, associated with changes in hormone therapy use in these 2 groups.
Collapse
Affiliation(s)
- Rowan T Chlebowski
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Aaron K Aragaki
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Garnet L Anderson
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ross L Prentice
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
256
|
Lello S, Capozzi A, Scambia G. Is there still a role for SERMs in menopause management? Gynecol Endocrinol 2020; 36:567-568. [PMID: 32530328 DOI: 10.1080/09513590.2020.1779691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Affiliation(s)
- Stefano Lello
- Department of Woman and Child Health, Policlinico Gemelli Foundation-IRCCS, Rome, Italy
| | - Anna Capozzi
- Department of Woman and Child Health, Policlinico Gemelli Foundation-IRCCS, Rome, Italy
| | - Giovanni Scambia
- Department of Woman and Child Health, Policlinico Gemelli Foundation-IRCCS, Rome, Italy
| |
Collapse
|
257
|
Oceguera-Basurto P, Topete A, Oceguera-Villanueva A, Rivas-Carrillo J, Paz-Davalos M, Quintero-Ramos A, Del Toro-Arreola A, Daneri-Navarro A. Selective estrogen receptor modulators in the prevention of breast cancer in premenopausal women: a review. Transl Cancer Res 2020; 9:4444-4456. [PMID: 35117809 PMCID: PMC8797886 DOI: 10.21037/tcr-19-1956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 06/15/2020] [Indexed: 12/28/2022]
Abstract
The detection of premenopausal women at high risk of breast cancer is key to chemoprevention. Therapy with selective estrogen receptor modulators (SERMs) induces a significant antiproliferative effect in estrogen receptor (ER) positive breast cancer. This review was designed according the guidelines of the 2009 PRISMA statement. Searching different databases, including PubMed, MedlinePlus, PLoS One, Cochrane Breast Cancer Specialized Register, Clinical Trials.gov and American Society of Clinical Oncology. From 168 records screened, 15 full text articles were assessed for eligibility and only 7 studies met the inclusion criteria. Three of the studies included analyzed changes in Ki-67 expression, revealing weaker expression after treatment with acolbifene and raloxifene (P<0.001). Three studies also analyzed the breast volume by magnetic resonance imagining (MRI) and demonstrate a significant difference after 1 year with raloxifene treatment (P=0.0017). Moreover, a 20% reduction in breast density was observed after a 2-year treatment with tamoxifen in premenopausal women. SERMs reduce the risk of developing breast cancer. The studies reviewed here demonstrate the modulation of Ki-67 expression and changes in breast density, suggesting an important preventive role for this group of drugs in prevention for premenopausal women at high risk of developing breast cancer.
Collapse
Affiliation(s)
- Paola Oceguera-Basurto
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, México
| | - Antonio Topete
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, México
| | | | - Jorge Rivas-Carrillo
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, México
| | | | - Antonio Quintero-Ramos
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, México
| | - Alicia Del Toro-Arreola
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, México
| | - Adrián Daneri-Navarro
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, México
| |
Collapse
|
258
|
Goemans N, Wong B, Van den Hauwe M, Signorovitch J, Sajeev G, Cox D, Landry J, Jenkins M, Dieye I, Yao Z, Hossain I, Ward SJ, the Collaborative Trajectory Analysis Project (cTAP). Prognostic factors for changes in the timed 4-stair climb in patients with Duchenne muscular dystrophy, and implications for measuring drug efficacy: A multi-institutional collaboration. PLoS One 2020; 15:e0232870. [PMID: 32555695 PMCID: PMC7302444 DOI: 10.1371/journal.pone.0232870] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 04/22/2020] [Indexed: 12/25/2022] Open
Abstract
The timed 4-stair climb (4SC) assessment has been used to measure function in Duchenne muscular dystrophy (DMD) practice and research. We sought to identify prognostic factors for changes in 4SC, assess their consistency across data sources, and the extent to which prognostic scores could be useful in DMD clinical trial design and analysis. Data from patients with DMD in the placebo arm of a phase 3 trial (Tadalafil DMD trial) and two real-world sources (Universitaire Ziekenhuizen, Leuven, Belgium [Leuven] and Cincinnati Children's Hospital Medical Center [CCHMC]) were analyzed. One-year changes in 4SC completion time and velocity (stairs/second) were analyzed. Prognostic models included age, height, weight, steroid use, and multiple timed function tests and were developed using multivariable regression, separately in each data source. Simulations were used to quantify impacts on trial sample size requirements. Data on 1-year changes in 4SC were available from the Tadalafil DMD trial (n = 92) Leuven (n = 67), and CCHMC (n = 212). Models incorporating multiple timed function tests, height, and weight significantly improved prognostic accuracy for 1-year change in 4SC (R2: 29%-36% for 4SC velocity, and 29%-34% for 4SC time) compared to models including only age, baseline 4SC and steroid duration (R2:8%-17% for 4SC velocity and 2%-13% for 4SC time). Measures of walking and rising ability contributed important prognostic information for changes in 4SC. In a randomized trial with equal allocation to treatment and placebo, adjustment for such a prognostic score would enable detection (at 80% power) of a treatment effect of 0.25 stairs/second with 100–120 patients, compared to 170–190 patients without prognostic score adjustment. Combining measures of ambulatory function doubled prognostic accuracy for 1-year changes in 4SC completion time and velocity. Randomized clinical trials incorporating a validated prognostic score could reduce sample size requirements by approximately 40%. Knowledge of important prognostic factors can also inform adjusted comparisons to external controls.
Collapse
Affiliation(s)
- Nathalie Goemans
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
- * E-mail:
| | - Brenda Wong
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, United States of America
| | | | - James Signorovitch
- Analysis Group Inc., Boston, Massachusetts, United States of America
- The Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
| | - Gautam Sajeev
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - David Cox
- Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - John Landry
- Eli Lilly and Company, Toronto, Ontario, Canada
| | | | - Ibrahima Dieye
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Zhiwen Yao
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Intekhab Hossain
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Susan J. Ward
- The Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
| | | |
Collapse
|
259
|
Zayas J, Qin S, Yu J, Ingle JN, Wang L. Functional genomics based on germline genome-wide association studies of endocrine therapy for breast cancer. Pharmacogenomics 2020; 21:615-625. [PMID: 32539536 DOI: 10.2217/pgs-2019-0191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Breast cancer is the most common invasive cancer in women worldwide. Functional follow-up of breast cancer genome-wide association studies has led to the discovery of genes that regulate endocrine therapy response in a SNP- and drug-dependent manner. Here, we will present four examples in which functional genomic studies from breast cancer clinical trials led to novel pharmacogenomic insights and molecular mechanisms of selective estrogen receptor modulators and aromatase inhibitors. The approach utilized for studying genetic variability described in this review offers substantial potential for meaningful discoveries that move the field toward precision medicine for patients.
Collapse
Affiliation(s)
- Jacqueline Zayas
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic School of Medicine & Mayo Clinic Medical Scientist Training Program, Rochester, MN 55905, USA
| | - Sisi Qin
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Jia Yu
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Liewei Wang
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
260
|
Jannati M, Ardecani AA. Prevention of Pulmonary and Venous Thromboembolism Post Coronary Artery Bypass Graft Surgery - Literature Review. Braz J Cardiovasc Surg 2020; 35:368-374. [PMID: 32549108 PMCID: PMC7299585 DOI: 10.21470/1678-9741-2018-0345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Objective The current review evaluates recent literature on the different aspects of prophylaxis in postoperative pulmonary and venous thromboembolism and their main risk factors. Methods The literature survey was carried out based on the PubMed data using the keywords “coronary artery bypass graft” and “venous thromboembolism” as components of the search field title. Results Studies reported several risk factors for postoperative thromboembolism including advanced age, postoperative immobilization, type of thromboprophylaxis, obesity, and location of the surgery. Conclusion According to the studies, tailored prophylaxis could be easily adapted to decrease the intensity and duration of postoperative thromboembolism in a patient with several disorders and comorbidities, especially in cardiovascular disease.
Collapse
Affiliation(s)
- Mansour Jannati
- Shiraz University of Medical Sciences Faghihi Hospital Department of Cardiovascular Surgery Shiraz Iran Department of Cardiovascular Surgery, Faghihi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Abdi Ardecani
- Shiraz University of Medical Sciences Faghihi Hospital Department of Cardiovascular Surgery Shiraz Iran Department of Cardiovascular Surgery, Faghihi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
261
|
Chelmow D, Pearlman MD, Young A, Bozzuto L, Dayaratna S, Jeudy M, Kremer ME, Scott DM, O'Hara JS. Executive Summary of the Early-Onset Breast Cancer Evidence Review Conference. Obstet Gynecol 2020; 135:1457-1478. [PMID: 32459439 PMCID: PMC7253192 DOI: 10.1097/aog.0000000000003889] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/23/2020] [Accepted: 03/12/2020] [Indexed: 12/23/2022]
Abstract
The Centers for Disease Control and Prevention launched the Bring Your Brave campaign to increase knowledge about early-onset breast cancer, defined as breast cancer in women aged 18-45 years. The American College of Obstetricians and Gynecologists convened a panel of experts in breast disease from the Society for Academic Specialists in General Obstetrics and Gynecology to review relevant literature, validated tools, best practices, and practice guidelines as a first step toward developing educational materials for women's health care providers about early-onset breast cancer. Panel members conducted structured literature reviews, which were then reviewed by other panel members and discussed at an in-person meeting of stakeholder professional and patient advocacy organizations in April 2019. This article summarizes the relevant literature, existing guidance, and validated tools to guide health care providers in the prevention, early detection, and special considerations of early-onset breast cancer. Substantive knowledge gaps were noted and summarized to provide guidance for future research.
Collapse
Affiliation(s)
- David Chelmow
- Department of Obstetrics and Gynecology, Virginia Commonwealth University School of Medicine, Richmond, Virginia; the Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, Michigan; the Department of Women's Health, the University of Texas at Austin Dell Medical School, Austin, Texas; the Departments of Obstetrics and Gynecology and Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; the Department of Obstetrics and Gynecology, Thomas Jefferson University Hospital, Sidney Kimmel Medical College, Philadelphia, Pennsylvania; Southeast Kaiser Permanente Medical Group, Atlanta, Georgia; the Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington; the Department of Obstetrics and Gynecology, University of Connecticut Medical School, Farmington, Connecticut; and the American College of Obstetricians and Gynecologists, Washington, DC
| | | | | | | | | | | | | | | | | |
Collapse
|
262
|
Biernacki K, Daśko M, Ciupak O, Kubiński K, Rachon J, Demkowicz S. Novel 1,2,4-Oxadiazole Derivatives in Drug Discovery. Pharmaceuticals (Basel) 2020; 13:ph13060111. [PMID: 32485996 PMCID: PMC7345688 DOI: 10.3390/ph13060111] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
Five-membered 1,2,4-oxadiazole heterocyclic ring has received considerable attentionbecause of its unique bioisosteric properties and an unusually wide spectrum of biological activities.Thus, it is a perfect framework for the novel drug development. After a century since the1,2,4-oxadiazole have been discovered, the uncommon potential attracted medicinal chemists'attention, leading to the discovery of a few presently accessible drugs containing 1,2,4-oxadiazoleunit. It is worth noting that the interest in a 1,2,4-oxadiazoles' biological application has been doubledin the last fifteen years. Herein, after a concise historical introduction, we present a comprehensiveoverview of the recent achievements in the synthesis of 1,2,4-oxadiazole-based compounds and themajor advances in their biological applications in the period of the last five years as well as briefremarks on prospects for further development.
Collapse
Affiliation(s)
- Karol Biernacki
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland; (K.B.); (O.C.); (J.R.)
| | - Mateusz Daśko
- Department of Inorganic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland;
| | - Olga Ciupak
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland; (K.B.); (O.C.); (J.R.)
| | - Konrad Kubiński
- Department of Molecular Biology, Faculty of Biotechnology and Environment Sciences, The John Paul II Catholic University of Lublin, Konstantynów 1i, 20-708 Lublin, Poland;
| | - Janusz Rachon
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland; (K.B.); (O.C.); (J.R.)
| | - Sebastian Demkowicz
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland; (K.B.); (O.C.); (J.R.)
- Correspondence:
| |
Collapse
|
263
|
Nasiri Z, Montazeri H, Akbari N, Mirfazli SS, Tarighi P. Synergistic Cytotoxic and Apoptotic Effects of Local Probiotic Lactobacillus Brevis Isolated from Regional Dairy Products in Combination with Tamoxifen. Nutr Cancer 2020; 73:290-299. [DOI: 10.1080/01635581.2020.1743871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Zahra Nasiri
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Montazeri
- School of Pharmacy‐International Campus, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Akbari
- Department of Microbiology, Faculty of Science, Islamic Azad University, Arak, Iran
| | - Seyedeh Sara Mirfazli
- Department of Medical Chemistry, School of Pharmacy- International Campus, Iran University of Medical Sciences, Tehran, Iran
| | - Parastoo Tarighi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
264
|
Lazzeroni M, Puntoni M, Provinciali N, Webber TB, Briata IM, D'Amico M, Giuliano S, Siri G, Cagnacci S, DeCensi A. Estimating the magnitude of clinical benefit of systemic therapy in patients with DCIS or pre-invasive disease of the breast. Breast 2020; 48 Suppl 1:S39-S43. [PMID: 31839158 DOI: 10.1016/s0960-9776(19)31121-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The challenge of effective management of ductal carcinoma in situ (DCIS) and other pre-malignant disorders of the breast is to select patients who will not progress to invasive carcinoma from those at the highest risk who require radiotherapy and/or endocrine therapy to minimize the risk of a subsequent invasive recurrence. Although IBIS-II and NSABP-B35 DCIS phase III trials proved that tamoxifen 20 mg/day and anastrozole reduce the risk of ipsilateral and contralateral events, the toxicities of both drugs have hampered the drug uptake by high-risk women. We recently reported results of a 3-year placebo-controlled trial of low-dose (5 mg/d) tamoxifen in 500 women with intraepithelial neoplasia (70% DCIS). At a median follow-up of 5 years, women randomly assigned to low-dose tamoxifen had half the number of subsequent diagnoses of DCIS or invasive cancer compared with those randomly assigned to placebo but no increase in thromboembolic events or endometrial cancers. The 5-year number needed to treat was 22 (95% CI, 20-27). Our attention is now focused on prognostic and predictive markers to identify patients who can derive the greatest benefits from low dose tamoxifen, such as for instance the expression of 23 genes involved in cell cycle progression (CCP). In conclusion, we endorse an active treatment of DCIS as the standard of care.
Collapse
Affiliation(s)
- Matteo Lazzeroni
- Division of Cancer Prevention and Genetics, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Matteo Puntoni
- Clinical Trial Office, E.O. Ospedali Galliera, Genoa, Italy
| | | | | | | | - Mauro D'Amico
- Division of Medical Oncology, E.O. Ospedali Galliera, Genoa, Italy
| | - Silvia Giuliano
- Division of Medical Oncology, E.O. Ospedali Galliera, Genoa, Italy
| | - Giacomo Siri
- Clinical Trial Office, E.O. Ospedali Galliera, Genoa, Italy
| | - Sara Cagnacci
- Division of Cancer Prevention and Genetics, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Andrea DeCensi
- Division of Medical Oncology, E.O. Ospedali Galliera, Genoa, Italy; Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom.
| |
Collapse
|
265
|
Current and future approaches to screening for endometrial cancer. Best Pract Res Clin Obstet Gynaecol 2020; 65:79-97. [DOI: 10.1016/j.bpobgyn.2019.12.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023]
|
266
|
Recommendations for prioritization, treatment, and triage of breast cancer patients during the COVID-19 pandemic. the COVID-19 pandemic breast cancer consortium. Breast Cancer Res Treat 2020; 181:487-497. [PMID: 32333293 PMCID: PMC7181102 DOI: 10.1007/s10549-020-05644-z] [Citation(s) in RCA: 271] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 12/19/2022]
Abstract
The COVID-19 pandemic presents clinicians a unique set of challenges in managing breast cancer (BC) patients. As hospital resources and staff become more limited during the COVID-19 pandemic, it becomes critically important to define which BC patients require more urgent care and which patients can wait for treatment until the pandemic is over. In this Special Communication, we use expert opinion of representatives from multiple cancer care organizations to categorize BC patients into priority levels (A, B, C) for urgency of care across all specialties. Additionally, we provide treatment recommendations for each of these patient scenarios. Priority A patients have conditions that are immediately life threatening or symptomatic requiring urgent treatment. Priority B patients have conditions that do not require immediate treatment but should start treatment before the pandemic is over. Priority C patients have conditions that can be safely deferred until the pandemic is over. The implementation of these recommendations for patient triage, which are based on the highest level available evidence, must be adapted to current availability of hospital resources and severity of the COVID-19 pandemic in each region of the country. Additionally, the risk of disease progression and worse outcomes for patients need to be weighed against the risk of patient and staff exposure to SARS CoV-2 (virus associated with the COVID-19 pandemic). Physicians should use these recommendations to prioritize care for their BC patients and adapt treatment recommendations to the local context at their hospital.
Collapse
|
267
|
Hwang ES, Hyslop T, Hendrix LH, Duong S, Bedrosian I, Price E, Caudle A, Hieken T, Guenther J, Hudis CA, Winer E, Lyss AP, Dickson-Witmer D, Hoefer R, Ollila DW, Hardman T, Marks J, Chen YY, Krings G, Esserman L, Hylton N. Phase II Single-Arm Study of Preoperative Letrozole for Estrogen Receptor-Positive Postmenopausal Ductal Carcinoma In Situ: CALGB 40903 (Alliance). J Clin Oncol 2020; 38:1284-1292. [PMID: 32125937 PMCID: PMC7164489 DOI: 10.1200/jco.19.00510] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2020] [Indexed: 01/31/2023] Open
Abstract
PURPOSE Primary endocrine therapy for ductal carcinoma in situ (DCIS) as a potential alternative to surgery has been understudied. This trial explored the feasibility of a short-term course of letrozole and sought to determine whether treatment results in measurable radiographic and biologic changes in estrogen receptor (ER)-positive DCIS. PATIENTS AND METHODS A phase II single-arm multicenter cooperative-group trial was conducted in postmenopausal patients diagnosed with ER-positive DCIS without invasion. Patients were treated with letrozole 2.5 mg per day for 6 months before surgery. Breast magnetic resonance imaging (MRI) was obtained at baseline, 3 months, and 6 months. The primary end point was change in 6-month MRI enhancement volume compared with baseline. RESULTS Overall, 79 patients were enrolled and 70 completed 6 months of letrozole. Of these, 67 patients had MRI data available for each timepoint. Baseline MRI volumes ranged from 0.004 to 26.3 cm3. Median reductions from baseline MRI volume (1.4 cm3) were 0.6 cm3 (61.0%) at 3 months (P < .001) and 0.8 cm3 (71.7%) at 6 months (P < .001). Consistent reductions were seen in median baseline ER H-score (228; median reduction, 15.0; P = .005), progesterone receptor H-score (15; median reduction, 85.0; P < .001), and Ki67 score (12%; median reduction, 6.3%; P = .007). Of the 59 patients who underwent surgery per study protocol, persistent DCIS remained in 50 patients (85%), invasive cancer was detected in six patients (10%), and no residual DCIS or invasive cancer was seen in nine patients (15%). CONCLUSIONS In a cohort of postmenopausal women with ER-positive DCIS, preoperative letrozole resulted in significant imaging and biomarker changes. These findings support future trials of extended endocrine therapy as primary nonoperative treatment of some DCIS.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/diagnostic imaging
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/surgery
- Carcinoma, Intraductal, Noninfiltrating/diagnostic imaging
- Carcinoma, Intraductal, Noninfiltrating/drug therapy
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/surgery
- Cohort Studies
- Female
- Humans
- Letrozole/therapeutic use
- Magnetic Resonance Imaging
- Mammography
- Neoadjuvant Therapy
- Postmenopause
- Preoperative Care/methods
- Receptors, Estrogen/metabolism
Collapse
Affiliation(s)
| | - Terry Hyslop
- Duke Cancer Institute, Duke University, Durham, NC
- Alliance Statistics and Data Center, Duke University, Durham, NC
| | - Laura H. Hendrix
- Duke Cancer Institute, Duke University, Durham, NC
- Alliance Statistics and Data Center, Duke University, Durham, NC
| | - Stephanie Duong
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN
| | | | - Elissa Price
- University of California, San Francisco, San Francisco, CA
| | | | - Tina Hieken
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN
| | | | | | - Eric Winer
- Dana-Farber/Partners Cancer Care, Boston, MA
| | | | | | | | | | | | | | - Yunn-Yi Chen
- University of California, San Francisco, San Francisco, CA
| | - Gregor Krings
- University of California, San Francisco, San Francisco, CA
| | - Laura Esserman
- University of California, San Francisco, San Francisco, CA
| | - Nola Hylton
- University of California, San Francisco, San Francisco, CA
| |
Collapse
|
268
|
Kumar N, Singh V, Mehta G. Assessment of common risk factors and validation of the Gail model for breast cancer: A hospital-based study from Western India. Tzu Chi Med J 2020; 32:362-366. [PMID: 33163382 PMCID: PMC7605293 DOI: 10.4103/tcmj.tcmj_171_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/14/2019] [Accepted: 12/03/2019] [Indexed: 11/04/2022] Open
Abstract
Objective Modified Gail Model is a noninvasive, easy to implement risk estimation tool for absolute breast cancer risk. It was developed with data collected from non African American females and further modified for African-American, the Hispanic, and Native American populations. The use of this model for population outside the US and European country is not yet validated. We evaluated the prevalent risk factors and the effectiveness of the Gail model for risk assessment in our local Indian population. Materials and Methods A retrospective analysis of a prospectively maintained database was conducted on patients treated between 2008 and 2013. Six hundred and fifty patients were included in each group. Six questions were taken as per the breast cancer risk assessment tool calculator. A value of over 1.67% was taken as a high risk for breast cancer development. Results The mean age of the participant was 50 ± 21.3 years in cases and 41 ± 16.4 years in controls. Age and age at first childbirth >30 years were found to be significant and associated with increased risk of breast carcinoma, but the age at menarche, family history, previous breast biopsy, and atypical hyperplasia was no significant. The Gail model was assessed, and sensitivity was 10.30% and 96.30% specificity for our population. Positive and negative predictive values were 73.62% and 51.77%. Conclusion Our study concluded that the Gail model is not an appropriate risk assessment tool for the population in its present form. For the future application of this model, we need to perform a bigger study with a higher sample size representing a maximum number of local variabilities in the Indian population.
Collapse
Affiliation(s)
- Naveen Kumar
- Department of General Surgery, Rabindra Nath Tagore Medical College and Hospital, Udaipur, Rajasthan, India
| | - Vinit Singh
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, India
| | - Garima Mehta
- Department of General Surgery, Rabindra Nath Tagore Medical College and Hospital, Udaipur, Rajasthan, India
| |
Collapse
|
269
|
Good M, Castro K, Denicoff A, Finnigan S, Parreco L, Germain DS. National Cancer Institute: Restructuring to Support the Clinical Trials of the Future. Semin Oncol Nurs 2020; 36:151003. [PMID: 32265163 DOI: 10.1016/j.soncn.2020.151003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES To describe the evolution and structure of the National Cancer Institute clinical trials programs, their notable accomplishments, nurses' roles in these accomplishments, and the essential role of nursing today and in the future. DATA SOURCES Manuscripts, government publications, websites, and professional communications. CONCLUSION Change is inevitable and a constant factor in the world of advancing science and clinical research. Nurses' contribution to research and evidence-based practice will continue to grow and is vital as the scientific landscape evolves. IMPLICATIONS FOR NURSING PRACTICE As the understanding of cancer biology increases and clinical trials evolve, nurses will need to remain key team members and leaders in National Cancer Institute Community Oncology Research Program and National Cancer Trials Network trials and their associated infrastructure.
Collapse
Affiliation(s)
- Marjorie Good
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD.
| | - Kathleen Castro
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD
| | - Andrea Denicoff
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD
| | - Shanda Finnigan
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD
| | - Linda Parreco
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD
| | - Diane St Germain
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD
| |
Collapse
|
270
|
Zacarías-Lara OJ, Méndez-Luna D, Martínez-Ruíz G, García-Sanchéz JR, Fragoso-Vázquez MJ, Bello M, Becerra-Martínez E, García-Vázquez JB, Correa-Basurto J. Synthesis and In Vitro Evaluation of Tetrahydroquinoline Derivatives as Antiproliferative Compounds of Breast Cancer via Targeting the GPER. Anticancer Agents Med Chem 2020; 19:760-771. [PMID: 30451119 DOI: 10.2174/1871520618666181119094144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/03/2018] [Accepted: 11/04/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Some reports have demonstrated the role of the G Protein-coupled Estrogen Receptor (GPER) in growth and proliferation of breast cancer cells. OBJECTIVE In an effort to develop new therapeutic strategies against breast cancer, we employed an in silico study to explore the binding modes of tetrahydroquinoline 2 and 4 to be compared with the reported ligands G1 and G1PABA. METHODS This study aimed to design and filter ligands by in silico studies determining their Lipinski's rule, toxicity and binding properties with GPER to achieve experimental assays as anti-proliferative compounds of breast cancer cell lines. RESULTS In silico studies suggest as promissory two tetrahydroquinoline 2 and 4 which contain a carboxyl group instead of the acetyl group (as is needed for G1 synthesis), which add low (2) and high hindrance (4) chemical moieties to explore the polar, hydrophobic and hindrance effects. Docking and molecular dynamics simulations of the target compounds were performed with GPER to explore their binding mode and free energy values. In addition, the target small molecules were synthesized and assayed in vitro using breast cancer cells (MCF-7 and MDA-MB-231). Experimental assays showed that compound 2 decreased cell proliferation, showing IC50 values of 50µM and 25µM after 72h of treatment of MCF-7 and MDA-MB-231 cell lines, respectively. Importantly, compound 2 showed a similar inhibitory effect on proliferation as G1 compound in MDA-MB-231 cells, suggesting that both ligands reach the GPER-binding site in a similar way, as was demonstrated through in silico studies. CONCLUSION A concentration-dependent inhibition of cell proliferation occurred with compound 2 in the two cell lines regardless of GPER.
Collapse
Affiliation(s)
- Oscar J Zacarías-Lara
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovacion Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron, 11340 Mexico, CDMX, Mexico
| | - David Méndez-Luna
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovacion Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron, 11340 Mexico, CDMX, Mexico
| | - Gustavo Martínez-Ruíz
- Unidad de Investigacion en Enfermedades Oncologicas, Hospital Infantil de Mexico, Federico Gomez, Mexico
| | - José R García-Sanchéz
- Laboratorio de Oncologia Molecular y Estres Oxidativo, Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron, 11340 Mexico, CDMX, Mexico
| | - Manuel J Fragoso-Vázquez
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovacion Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron, 11340 Mexico, CDMX, Mexico.,Departamento de Química Orgánica, Escuela Nacional de Ciencias, Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala, México, CDMX., 11340 México
| | - Martiniano Bello
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovacion Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron, 11340 Mexico, CDMX, Mexico
| | - Elvia Becerra-Martínez
- Laboratorio de RMN, Centro de Nanociencias y Micro y Nanotecnologias, Instituto Politecnico Nacional, Calle Luis Enrique Erro s/n, Unidad Profesional Adolfo Lopez Mateos, Gustavo A, Madero, 07738 Mexico, Ciudad de Mexico, Mexico
| | - Juan B García-Vázquez
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovacion Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron, 11340 Mexico, CDMX, Mexico
| | - José Correa-Basurto
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovacion Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron, 11340 Mexico, CDMX, Mexico
| |
Collapse
|
271
|
Day CM, Hickey SM, Song Y, Plush SE, Garg S. Novel Tamoxifen Nanoformulations for Improving Breast Cancer Treatment: Old Wine in New Bottles. Molecules 2020; 25:E1182. [PMID: 32151063 PMCID: PMC7179425 DOI: 10.3390/molecules25051182] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/24/2020] [Accepted: 03/01/2020] [Indexed: 12/15/2022] Open
Abstract
Breast cancer (BC) is one of the leading causes of death from cancer in women; second only to lung cancer. Tamoxifen (TAM) is a hydrophobic anticancer agent and a selective estrogen modulator (SERM), approved by the FDA for hormone therapy of BC. Despite having striking efficacy in BC therapy, concerns regarding the dose-dependent carcinogenicity of TAM still persist, restricting its therapeutic applications. Nanotechnology has emerged as one of the most important strategies to solve the issue of TAM toxicity, owing to the ability of nano-enabled-formulations to deliver smaller concentrations of TAM to cancer cells, over a longer period of time. Various TAM-containing-nanosystems have been successfully fabricated to selectively deliver TAM to specific molecular targets found on tumour membranes, reducing unwanted toxic effects. This review begins with an outline of breast cancer, the current treatment options and a history of how TAM has been used as a combatant of BC. A detailed discussion of various nanoformulation strategies used to deliver lower doses of TAM selectively to breast tumours will then follow. Finally, a commentary on future perspectives of TAM being employed as a targeting vector, to guide the delivery of other therapeutic and diagnostic agents selectively to breast tumours will be presented.
Collapse
Affiliation(s)
- Candace M. Day
- School of Pharmacy and Medical Sciences, University of South Australia, Cancer Research Institute, North Terrace, 5000 Adelaide, SA, Australia; (C.M.D.); (S.M.H.); (Y.S.)
| | - Shane M. Hickey
- School of Pharmacy and Medical Sciences, University of South Australia, Cancer Research Institute, North Terrace, 5000 Adelaide, SA, Australia; (C.M.D.); (S.M.H.); (Y.S.)
| | - Yunmei Song
- School of Pharmacy and Medical Sciences, University of South Australia, Cancer Research Institute, North Terrace, 5000 Adelaide, SA, Australia; (C.M.D.); (S.M.H.); (Y.S.)
| | - Sally E. Plush
- School of Pharmacy and Medical Sciences, University of South Australia, Cancer Research Institute, North Terrace, 5000 Adelaide, SA, Australia; (C.M.D.); (S.M.H.); (Y.S.)
- Future Industry Institute, University of South Australia, 5095 Mawson Lakes, SA, Australia
| | - Sanjay Garg
- School of Pharmacy and Medical Sciences, University of South Australia, Cancer Research Institute, North Terrace, 5000 Adelaide, SA, Australia; (C.M.D.); (S.M.H.); (Y.S.)
- Future Industry Institute, University of South Australia, 5095 Mawson Lakes, SA, Australia
| |
Collapse
|
272
|
Lubet RA, Heckman-Stoddard BM, Fox JT, Moeinpour F, Juliana MM, Shoemaker RH, Grubbs CJ. Use of Biomarker Modulation in Normal Mammary Epithelium as a Correlate for Efficacy of Chemopreventive Agents Against Chemically Induced Cancers. Cancer Prev Res (Phila) 2020; 13:283-290. [PMID: 31871222 PMCID: PMC7060128 DOI: 10.1158/1940-6207.capr-19-0318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/06/2019] [Accepted: 12/18/2019] [Indexed: 11/16/2022]
Abstract
In both estrogen receptor/progesterone receptor-positive (ER+/PR+) human breast cancer and in ER+/PR+ cancers in the methylnitrosourea (MNU)-induced rat model, short-term modulation of proliferation in early cancers predicts preventive/therapeutic efficacy. We determined the effects of known effective/ineffective chemopreventive agents on proliferative index (PI) in both rat mammary epithelium and small cancers. Female Sprague-Dawley rats were treated with MNU at 50 days of age. Five days later, the rats were treated with the individual compounds for a period of 14 days. At that time, normal mammary tissue from the inguinal gland area was surgically removed. After removal, the rats remained on the agents for an additional 5 months. This cancer prevention study confirmed our prior results of striking efficacy with tamoxifen, vorozole, Targretin, and gefitinib, and no efficacy with metformin, naproxen, and Lipitor. Employing a separate group of rats, the effects of short-term (7 days) drug exposure on small palpable cancers were examined. The PI in both small mammary cancers and in normal epithelium from control rats was >12%. In agreement with the cancer multiplicity data, tamoxifen, vorozole, gefitinib, and Targretin all strongly inhibited proliferation (>65%; P < 0.025) in the normal mammary epithelium. The ineffective agents metformin, naproxen, and Lipitor minimally affected PI. In the small cancers, tamoxifen, vorozole, and Targretin all reduced the PI, while metformin and Lipitor failed to do so. Thus, short-term changes in the PI in either normal mammary epithelium or small cancers correlated with long-term preventive efficacy in the MNU-induced rat model.
Collapse
Affiliation(s)
- Ronald A Lubet
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, NCI, Rockville, Maryland.
| | - Brandy M Heckman-Stoddard
- Breast and Gynecologic Cancer Research Group, Division of Cancer Prevention, NCI, Rockville, Maryland
| | - Jennifer T Fox
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, NCI, Rockville, Maryland
| | - Fariba Moeinpour
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - M Margaret Juliana
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Robert H Shoemaker
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, NCI, Rockville, Maryland
| | - Clinton J Grubbs
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
273
|
Kandagatla P, Rizk NN, Dokic D, Kochkodan J, Estevez S, Yanik M, Goranta S, Huber‐Keener K, Jeruss JS. Patient and provider factors associated with the noninitiation of tamoxifen for young women at high‐risk for the development of breast cancer. Breast J 2020; 26:464-468. [PMID: 31538708 PMCID: PMC10167625 DOI: 10.1111/tbj.13528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 01/31/2023]
Abstract
We sought to identify factors associated with disparities in tamoxifen utilization among young patients at high-risk for developing breast cancer. We identified 67 premenopausal, high-risk women age 35-45, without surgical prophylaxis, who did not initiate tamoxifen. Factors associated with noninitiation were examined. About 37% of patients had no documented provider-based discussion regarding initiation. Type of high-risk diagnosis was the only factor associated with a provider-based discussion (P = .03). For patients offered tamoxifen, primary reasons for noninitiation were perceived minimal benefit (66.7%), fertility concerns (16.7%), and concerns about side effects (7.1%). Implementation of comprehensive educational strategies regarding the benefits of tamoxifen should be facilitated to improve initiation among young high-risk patients.
Collapse
Affiliation(s)
- Pridvi Kandagatla
- Surgery University of Michigan Ann Arbor Michigan
- Surgery Henry Ford Hospital Detroit Michigan
| | | | | | | | | | - Megan Yanik
- University of Michigan School of Medicine Ann Arbor Michigan
| | - Sowmya Goranta
- Internal Medicine Michigan State University Flint Michigan
| | | | | |
Collapse
|
274
|
Bandera BC, Voci A, Nelson DW, Stern S, Barrak D, Fischer TD, DiNome ML, Goldfarb M. Disparities in Risk Reduction Therapy Recommendations for Young Women With Lobular Carcinoma In-Situ. Clin Breast Cancer 2020; 20:e397-e402. [PMID: 32081572 DOI: 10.1016/j.clbc.2020.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 10/19/2019] [Accepted: 01/17/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Endocrine therapy (ET) significantly reduces the risk of breast cancer development in high-risk patients diagnosed with lobular carcinoma in situ (LCIS). However, the variables impacting recommendation and use of ET in young adults (YAs) is not well-studied. We examined the role of provider recommendation and patient acceptance for ET for YAs with LCIS. MATERIALS AND METHODS The National Cancer Database was queried for women aged < 40 years with primary LCIS between 2000 and 2012. Socioeconomic, demographic, and treatment variables were examined to determine their impact on ET provider recommendation and initial patient acceptance of risk-reducing therapy. RESULTS Among 1650 YA patients with LCIS, only 749 (45.4%) were recommended ET. On multivariable analysis, women > 30 years of age were more likely recommended ET than women < 30 years (odds ratio [OR], 1.64; 95% confidence interval [CI], 1.10-2.47), African Americans more than other ethnicities (OR, 1.48; 95% CI, 1.1-2.0), and YAs treated in New England were more likely than those in the rest of the country (OR, 3.26; 95% CI, 2.0-5.2). Among YA women recommended ET, only 20.2% had a documented refusal. Only geography appeared to independently impact the likelihood of refusal, with YAs in the Southeastern-Central United States being most likely to refuse ET (OR, 5.4; 95% CI, 1.2-24.0). CONCLUSION ET is underutilized for risk-reduction in YAs with LCIS. This underuse appears dependent on disparities in provider recommendation practices rather than non-acceptance of therapy. This may reflect regional practice patterns, community standards of care, or provider bias regarding the significance of LCIS as a risk factor for development of invasive cancer.
Collapse
Affiliation(s)
- Bradley C Bandera
- Department of Surgery, John Wayne Cancer Institute, Santa Monica, CA; Present affiliation: Department of Surgery, Eisenhower Army Medical Center, Augusta, CA
| | - Amy Voci
- Department of Surgery, John Wayne Cancer Institute, Santa Monica, CA; Present affiliation: Department of Surgery, Carolina Medical Center, Charlotte, NC
| | - Daniel W Nelson
- Department of Surgery, John Wayne Cancer Institute, Santa Monica, CA; Present affiliation: Department of Surgery, William Beaumont Army Medical Center, El Paso, TX
| | - Stacey Stern
- Department of Surgery, John Wayne Cancer Institute, Santa Monica, CA
| | - Dany Barrak
- Department of Surgery, John Wayne Cancer Institute, Santa Monica, CA
| | - Trevan D Fischer
- Department of Surgery, John Wayne Cancer Institute, Santa Monica, CA
| | - Maggie L DiNome
- Department of Surgery, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA
| | - Melanie Goldfarb
- Department of Surgery, John Wayne Cancer Institute, Santa Monica, CA.
| |
Collapse
|
275
|
The Application of Gail Model to Predict the Risk of Developing Breast Cancer among Jordanian Women. JOURNAL OF ONCOLOGY 2020; 2020:9608910. [PMID: 32148498 PMCID: PMC7053471 DOI: 10.1155/2020/9608910] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/30/2019] [Accepted: 02/03/2020] [Indexed: 11/30/2022]
Abstract
Background and Objectives. Breast cancer has been the most common cancer affecting women in Jordan. In the process of implementing breast cancer prevention and early detection programs, individualized risk assessment can add to the cost-effectiveness of such interventions. Gail model is a widely used tool to stratify patients into different risk categories. However, concerns about its applicability across different ethnic groups do exist. In this study, we report our experience with the application of a modified version of this model among Jordanian women.
Collapse
|
276
|
Miller EA, Pinsky PF, Heckman-Stoddard BM, Minasian LM. Breast cancer risk prediction models and subsequent tumor characteristics. Breast Cancer 2020; 27:662-669. [PMID: 32056079 DOI: 10.1007/s12282-020-01060-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 02/05/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND A previous study found evidence that a breast cancer risk prediction model preferentially selected for less aggressive tumors in Swedish women. In the US, the Gail model has been widely used and was used for entry criteria in two large breast cancer prevention trials. We assessed if higher risk levels from the Gail model were associated with less aggressive tumor characteristics and if risk levels were predictive of mortality and survival. METHODS We used questionnaire data from women in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial to calculate Gail risk levels (low < 1.66%; moderate 1.66-2.99%; high ≥ 3.00%). Women aged 55-74 were enrolled between 1993 and 2001 and had detailed information on breast cancer incidence and tumors collected. We calculated breast cancer incidence and mortality rates among all women by risk levels and examined breast cancer survival and tumor characteristics among women diagnosed with breast cancer. We used Chi-squared tests and multivariable logistic regression to assess the association between risk levels and tumor characteristics. RESULTS The study population for this analysis included 45,402 women with 1908 cases of breast cancer. Women at high risk were associated with higher risk of breast cancer mortality compared to women with low risk [rate ratio (RR) = 2.29 95% confidence interval (CI) 1.37-3.84)]. Higher risk levels were associated with lobular-type tumors [moderate: adjusted odds ratio (aOR) = 1.57 95% CI 1.13-2.17; high: aOR = 1.78 95% CI 1.25-2.54] but were not associated with any other tumor characteristics or breast cancer survival. CONCLUSIONS We did not find evidence that higher risk levels from the Gail model are predictive of less aggressive breast cancer tumors.
Collapse
Affiliation(s)
- Eric A Miller
- Division of Cancer Prevention, National Cancer Institute, 9609 Medical Center Drive, Rockville, MD, 20850, USA.
| | - Paul F Pinsky
- Division of Cancer Prevention, National Cancer Institute, 9609 Medical Center Drive, Rockville, MD, 20850, USA
| | - Brandy M Heckman-Stoddard
- Division of Cancer Prevention, National Cancer Institute, 9609 Medical Center Drive, Rockville, MD, 20850, USA
| | - Lori M Minasian
- Division of Cancer Prevention, National Cancer Institute, 9609 Medical Center Drive, Rockville, MD, 20850, USA
| |
Collapse
|
277
|
Vogel VG. Breast cancer risk reduction: Notable achievements and remaining challenges. Breast J 2020; 26:86-91. [PMID: 31971344 DOI: 10.1111/tbj.13717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 10/04/2019] [Indexed: 12/26/2022]
Abstract
Millions of women in the United States are at increased risk of breast cancer. Multiple prospective, randomized clinical trials have demonstrated both the efficacy and safety of selective estrogen receptor modulators and aromatase inhibitors in reducing substantially the risk of invasive breast cancer in women at increased risk. Published tables are available to aid clinicians in shared decision-making regarding drug interventions with their patients who are at increased risk of breast cancer. Both professional and governmental agencies have advised that these interventions should be offered to women at increased risk of breast cancer. Doing so would reduce breast cancer morbidity substantially.
Collapse
Affiliation(s)
- Victor G Vogel
- Breast Medical Oncology/Research, Geisinger Health System, Danville, Pennsylvania
| |
Collapse
|
278
|
Affiliation(s)
- Michael A. Biersmith
- Cardio‐Oncology ProgramDivision of Cardiovascular MedicineDepartment of MedicineThe Ohio State UniversityColumbusOH
| | - Matthew S. Tong
- Cardio‐Oncology ProgramDivision of Cardiovascular MedicineDepartment of MedicineThe Ohio State UniversityColumbusOH
| | - Avirup Guha
- Cardio‐Oncology ProgramDivision of Cardiovascular MedicineDepartment of MedicineThe Ohio State UniversityColumbusOH
- Harrington Heart and Vascular InstituteCase Western Reserve UniversityClevelandOH
| | - Orlando P. Simonetti
- Cardio‐Oncology ProgramDivision of Cardiovascular MedicineDepartment of MedicineThe Ohio State UniversityColumbusOH
| | - Daniel Addison
- Cardio‐Oncology ProgramDivision of Cardiovascular MedicineDepartment of MedicineThe Ohio State UniversityColumbusOH
- Division of Cancer Prevention and ControlDepartment of MedicineCollege of MedicineThe Ohio State UniversityColumbusOH
| |
Collapse
|
279
|
Bueno MJ, Quintela-Fandino M. Emerging role of Fatty acid synthase in tumor initiation: implications for cancer prevention. Mol Cell Oncol 2020; 7:1709389. [PMID: 32158923 PMCID: PMC7051128 DOI: 10.1080/23723556.2019.1709389] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 11/24/2022]
Abstract
Targeting metabolic reprogramming has emerged as a promising strategy for therapeutic intervention in cancer. We identify that fatty acid synthase (FASN) is essential for cancer initiation playing a critical role in acquiring three-dimensional (3D) growth properties during transformation. In vivo inhibition of FASN before oncogenic activation prevents tumor development and invasive growth suggesting that FASN could be a potential target for cancer prevention.
Collapse
Affiliation(s)
- Maria J Bueno
- Breast Cancer Clinical Research Unit, CNIO-Spanish National Cancer Research Center, Madrid, Spain
| | - Miguel Quintela-Fandino
- Breast Cancer Clinical Research Unit, CNIO-Spanish National Cancer Research Center, Madrid, Spain.,Medical Oncology, Hospital Universitario Quiron, Pozuelo de Alarcon, Madrid, Spain.,Medical Oncology, Hospital Universitario de Fuenlabrada, Fuenlabrada, Madrid, Spain
| |
Collapse
|
280
|
Catalá-López F, Aleixandre-Benavent R, Caulley L, Hutton B, Tabarés-Seisdedos R, Moher D, Alonso-Arroyo A. Global mapping of randomised trials related articles published in high-impact-factor medical journals: a cross-sectional analysis. Trials 2020; 21:34. [PMID: 31910857 PMCID: PMC6947860 DOI: 10.1186/s13063-019-3944-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022] Open
Abstract
Background Randomised controlled trials (RCTs) provide the most reliable information to inform clinical practice and patient care. We aimed to map global clinical research publication activity through RCT-related articles in high-impact-factor medical journals over the past five decades. Methods We conducted a cross-sectional analysis of articles published in the highest ranked medical journals with an impact factor > 10 (according to Journal Citation Reports published in 2017). We searched PubMed/MEDLINE (from inception to December 31, 2017) for all RCT-related articles (e.g. primary RCTs, secondary analyses and methodology papers) published in high-impact-factor medical journals. For each included article, raw metadata were abstracted from the Web of Science. A process of standardization was conducted to unify the different terms and grammatical variants and to remove typographical, transcription and/or indexing errors. Descriptive analyses were conducted (including the number of articles, citations, most prolific authors, countries, journals, funding sources and keywords). Network analyses of collaborations between countries and co-words are presented. Results We included 39,305 articles (for the period 1965–2017) published in forty journals. The Lancet (n = 3593; 9.1%), the Journal of Clinical Oncology (n = 3343; 8.5%) and The New England Journal of Medicine (n = 3275 articles; 8.3%) published the largest number of RCTs. A total of 154 countries were involved in the production of articles. The global productivity ranking was led by the United States (n = 18,393 articles), followed by the United Kingdom (n = 8028 articles), Canada (n = 4548 articles) and Germany (n = 4415 articles). Seventeen authors who had published 100 or more articles were identified; the most prolific authors were affiliated with Duke University (United States), Harvard University (United States) and McMaster University (Canada). The main funding institutions were the National Institutes of Health (United States), Hoffmann-La Roche (Switzerland), Pfizer (United States), Merck Sharp & Dohme (United States) and Novartis (Switzerland). The 100 most cited RCTs were published in nine journals, led by The New England Journal of Medicine (n = 78 articles), The Lancet (n = 9 articles) and JAMA (n = 7 articles). These landmark contributions focused on novel methodological approaches (e.g. the “Bland-Altman method”) and trials on the management of chronic conditions (e.g. diabetes control, hormone replacement therapy in postmenopausal women, multiple therapies for diverse cancers, cardiovascular therapies such as lipid-lowering statins, antihypertensive medications, and antiplatelet and antithrombotic therapy). Conclusions Our analysis identified authors, countries, funding institutions, landmark contributions and high-impact-factor medical journals publishing RCTs. Over the last 50 years, publication production in leading medical journals has increased, with Western countries leading in research but with low- and middle-income countries showing very limited representation.
Collapse
Affiliation(s)
- Ferrán Catalá-López
- Department of Health Planning and Economics, National School of Public Health, Institute of Health Carlos III, Madrid, Spain. .,Department of Medicine, University of Valencia/INCLIVA Health Research Institute and CIBERSAM, Valencia, Spain. .,Clinical Epidemiology Program, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada.
| | - Rafael Aleixandre-Benavent
- Ingenio-Spanish National Research Council (CSIC) and Universitat Politècnica de Valencia (UPV), Valencia, Spain.,Information and Social and Health Research Unit (UISYS), University of Valencia and Spanish National Research Council (CSIC), Valencia, Spain
| | - Lisa Caulley
- Clinical Epidemiology Program, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada.,Ear, Nose and Throat Department, Guy's Hospital, London, UK.,Department of Clinical Epidemiology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Brian Hutton
- Clinical Epidemiology Program, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Rafael Tabarés-Seisdedos
- Department of Medicine, University of Valencia/INCLIVA Health Research Institute and CIBERSAM, Valencia, Spain
| | - David Moher
- Clinical Epidemiology Program, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Adolfo Alonso-Arroyo
- Information and Social and Health Research Unit (UISYS), University of Valencia and Spanish National Research Council (CSIC), Valencia, Spain.,Department of History of Science and Documentation, University of Valencia, Valencia, Spain
| |
Collapse
|
281
|
Minami CA, Zabor EC, Gilbert E, Newman A, Park A, Jochelson MS, King TA, Pilewskie ML. Do Body Mass Index and Breast Density Impact Cancer Risk Among Women with Lobular Carcinoma In Situ? Ann Surg Oncol 2020; 27:1844-1851. [PMID: 31898097 DOI: 10.1245/s10434-019-08126-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Indexed: 11/18/2022]
Abstract
PURPOSE Both body mass index (BMI) and breast density impact breast cancer risk in the general population. Whether obesity and density represent additive risk factors in women with lobular carcinoma in situ (LCIS) is unknown. METHODS Patients diagnosed with LCIS from 1988 to 2017 were identified from a prospectively maintained database. BMI was categorized by World Health Organization classification. Density was captured as the mammographic Breast Imaging Reporting and Data System (BIRADS) value. Other covariates included age at LCIS diagnosis, menopausal status, family history, chemoprevention, and prophylactic mastectomy. Cancer-free probability was estimated using the Kaplan-Meier method, and Cox regression models were used for univariable and multivariable analyses. RESULTS A total of 1222 women with LCIS were identified. At a median follow-up of 7 years, 179 women developed breast cancer (121 invasive, 58 ductal carcinoma in situ); 5- and 10-year cumulative incidences of breast cancer were 10% and 17%, respectively. In multivariable analysis, increased breast density (BIRADS C/D vs. A/B) was significantly associated with increased hazard of breast cancer (hazard ratio [HR] 2.42, 95% confidence interval [CI] 1.52-3.88), whereas BMI was not. On multivariable analysis, chemoprevention use was associated with a significantly decreased hazard of breast cancer (HR 0.49, 95% CI 0.29-0.84). Exploratory analyses did not demonstrate significant interaction between BMI and menopausal status, BMI and breast density, BMI and chemoprevention use, or breast density and chemoprevention. CONCLUSIONS Breast cancer risk among women with LCIS is impacted by breast density. These results aid in personalizing risk assessment among women with LCIS and highlight the importance of chemoprevention counseling for risk reduction.
Collapse
Affiliation(s)
- Christina A Minami
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA.,Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA
| | - Emily C Zabor
- Biostatistics Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Ashley Newman
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anna Park
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maxine S Jochelson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tari A King
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA.,Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA
| | - Melissa L Pilewskie
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
282
|
Tumors of the Female Reproductive Organs. Fam Med 2020. [DOI: 10.1007/978-1-4939-0779-3_112-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
283
|
Basen-Engquist K, Brown P, Coletta AM, Savage M, Maresso KC, Hawk E. Lifestyle and Cancer Prevention. ABELOFF'S CLINICAL ONCOLOGY 2020:337-374.e12. [DOI: 10.1016/b978-0-323-47674-4.00022-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
284
|
Abnormal Screening Mammogram. Surgery 2020. [DOI: 10.1007/978-3-030-05387-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
285
|
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
|
286
|
Liu W, Qdaisat A, Lopez G, Narayanan S, Underwood S, Spano M, Reddy A, Guo Y, Zhou S, Yeung SC, Bruera E, Garcia MK, Cohen L. Acupuncture for Hot Flashes in Cancer Patients: Clinical Characteristics and Traditional Chinese Medicine Diagnosis as Predictors of Treatment Response. Integr Cancer Ther 2019; 18:1534735419848494. [PMID: 31046489 PMCID: PMC6501481 DOI: 10.1177/1534735419848494] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Acupuncture is a recognized integrative modality for managing hot flashes. However, data regarding predictors for response to acupuncture in cancer patients experiencing hot flashes are limited. We explored associations between patient characteristics, including traditional Chinese medicine (TCM) diagnosis, and treatment response among cancer patients who received acupuncture for management of hot flashes. Methods: We reviewed acupuncture records of cancer outpatients with the primary reason for referral listed as hot flashes who were treated from March 2016 to April 2018. Treatment response was assessed using the hot flashes score within a modified Edmonton Symptom Assessment Scale (0-10 scale) administered immediately before and after each acupuncture treatment. Correlations between TCM diagnosis, individual patient characteristics, and treatment response were analyzed. Results: The final analysis included 558 acupuncture records (151 patients). The majority of patients were female (90%), and 66% had breast cancer. The median treatment response was a 25% reduction in the hot flashes score. The most frequent TCM diagnosis was qi stagnation (80%) followed by blood stagnation (57%). Older age (P = .018), patient self-reported anxiety level (P = .056), and presence of damp accumulation in TCM diagnosis (P = .047) were correlated with greater hot flashes score reduction. Conclusions: TCM diagnosis and other patient characteristics were predictors of treatment response to acupuncture for hot flashes in cancer patients. Future research is needed to further explore predictors that could help tailor acupuncture treatments for these patients.
Collapse
Affiliation(s)
- Wenli Liu
- 1 University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aiham Qdaisat
- 1 University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel Lopez
- 1 University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Susan Underwood
- 1 University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Spano
- 1 University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Akhila Reddy
- 1 University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ying Guo
- 1 University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shouhao Zhou
- 2 Pennsylvania State University, Hershey, PA, USA
| | - Sai-Ching Yeung
- 1 University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eduardo Bruera
- 1 University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Kay Garcia
- 1 University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lorenzo Cohen
- 1 University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
287
|
Ball S, Arevalo M, Juarez E, Payne JD, Jones C. Breast cancer chemoprevention: An update on current practice and opportunities for primary care physicians. Prev Med 2019; 129:105834. [PMID: 31494144 DOI: 10.1016/j.ypmed.2019.105834] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 09/01/2019] [Accepted: 09/05/2019] [Indexed: 12/15/2022]
Abstract
Several risk assessment models have been validated for the estimation of risk of breast cancer in women. Chemoprevention through hormonal therapy is an effective way to reduce the incidence of breast cancer in women with high risk. Selective estrogen receptor modulators, tamoxifen and raloxifene, are approved for this indication by the United States Food and Drug Administration, and aromatase inhibitors have also shown promise in recent studies. These medications are generally well tolerated, except for reported increased rates of fractures and venous thromboembolic events. Despite strong recommendations from several regulatory bodies, advocacy for chemoprevention has been inadequate in practice, more so among the primary care physicians. Studies have identified several barriers in physicians, patients, and the system, contributing to this problem. Lack of knowledge about risk assessment models and chemoprevention options preclude physicians from prescribing these medications with confidence. Fear of potential adverse events, confusion regarding the purpose of the therapy, and need for continued adherence for five years are among the principal reasons for reduced chemoprevention uptake and early discontinuation among patients. Multifaceted interventions directed at education and training of health care professionals, proper counseling of women at high risk, and promotion of the development of improved medications might help ensure better chemoprevention uptake in the target population.
Collapse
Affiliation(s)
- Somedeb Ball
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Meily Arevalo
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Edna Juarez
- Department of Internal Medicine, Memorial Medical Center, Las Cruces, NM, USA
| | - J Drew Payne
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Catherine Jones
- Division of Hematology and Medical Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
288
|
Crew KD, Silverman TB, Vanegas A, Trivedi MS, Dimond J, Mata J, Sin M, Jones T, Terry MB, Tsai WY, Kukafka R. Study protocol: Randomized controlled trial of web-based decision support tools for high-risk women and healthcare providers to increase breast cancer chemoprevention. Contemp Clin Trials Commun 2019; 16:100433. [PMID: 31497674 PMCID: PMC6722284 DOI: 10.1016/j.conctc.2019.100433] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/11/2019] [Accepted: 08/19/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Chemoprevention using selective estrogen receptor modulators and aromatase inhibitors has been shown to reduce invasive breast cancer incidence in high-risk women. Despite this evidence, few high-risk women who are eligible for chemoprevention utilize it as a risk-reducing strategy. Reasons for low uptake include inadequate knowledge about chemoprevention among patients and healthcare providers, concerns about side effects, time constraints during the clinical encounter, and competing comorbidities. METHODS/DESIGN We describe the study design of a randomized controlled trial examining the effect of two web-based decision support tools on chemoprevention decision antecedents and quality, referral for specialized counseling, and chemoprevention uptake among women at an increased risk for breast cancer. The trial is being conducted at a large, urban medical center. A total of 300 patients and 50 healthcare providers will be recruited and randomized to standard educational materials alone or in combination with the decision support tools. Patient reported outcomes will be assessed at baseline, one and six months after randomization, and after their clinic visit with their healthcare provider. DISCUSSION We are conducting this trial to provide evidence on how best to support personalized breast cancer risk assessment and informed and shared decision-making for chemoprevention. We propose to integrate the decision support tools into clinical workflow, which can potentially expand quality decision-making and chemoprevention uptake. TRIAL REGISTRATION NCT03069742.
Collapse
Affiliation(s)
- Katherine D. Crew
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Thomas B. Silverman
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Alejandro Vanegas
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Meghna S. Trivedi
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Jill Dimond
- Sassafras Tech Collective, Ann Arbor, MI, USA
| | - Jennie Mata
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Margaret Sin
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Tarsha Jones
- Christine E Lynn College of Nursing, Florida Atlantic University, Boca Raton, FL, USA
| | - Mary Beth Terry
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Wei-Yann Tsai
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Rita Kukafka
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Sociomedical Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
289
|
Fricker J. Bernard Fisher: US surgeon who pioneered a scientific approach for breast cancer treatment. Assoc Med J 2019. [DOI: 10.1136/bmj.l6657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
290
|
Thorat MA, Balasubramanian R. Breast cancer prevention in high-risk women. Best Pract Res Clin Obstet Gynaecol 2019; 65:18-31. [PMID: 31862315 DOI: 10.1016/j.bpobgyn.2019.11.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/10/2019] [Accepted: 11/11/2019] [Indexed: 12/24/2022]
Abstract
Women at high risk of developing breast cancer are a heterogeneous group of women including those with and without high-risk germline mutation/s. Prevention in these women requires a personalised and multidisciplinary approach. Preventive therapy with selective oestrogen receptor modulators (SERMs) like tamoxifen and aromatase inhibitors (AIs) substantially reduces breast cancer risk well beyond the active treatment period. The importance of benign breast disease as a marker of increased breast cancer risk remains underappreciated, and although the benefit of preventive therapy may be greater in such women, preventive therapy remains underutilised in these and other high-risk women. Bilateral Risk-Reducing Mastectomy (BRRM) reduces the risk of developing breast cancer by 90% in high-risk women such as carriers of BRCA mutations. It also improves breast cancer-specific survival in BRCA1 carriers. Bilateral risk-reducing salpingo-oophorectomy may also reduce risk in premenopausal BRCA2 carriers. Further research to improve risk models, to identify surrogate biomarkers of preventive therapy benefit and to develop newer preventive agents is needed.
Collapse
Affiliation(s)
- Mangesh A Thorat
- Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, United Kingdom; School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, United Kingdom; Breast Services, Guy's Hospital, Great Maze Pond, London, SE1 9RT, United Kingdom.
| | | |
Collapse
|
291
|
Visvanathan K, Fabian CJ, Bantug E, Brewster AM, Davidson NE, DeCensi A, Floyd JD, Garber JE, Hofstatter EW, Khan SA, Katapodi MC, Pruthi S, Raab R, Runowicz CD, Somerfield MR. Use of Endocrine Therapy for Breast Cancer Risk Reduction: ASCO Clinical Practice Guideline Update. J Clin Oncol 2019; 37:3152-3165. [DOI: 10.1200/jco.19.01472] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE To update the ASCO guideline on pharmacologic interventions for breast cancer risk reduction and provide guidance on clinical issues that arise when deciding to use endocrine therapy for breast cancer risk reduction. METHODS An Expert Panel conducted targeted systematic literature reviews to identify new studies. RESULTS A randomized clinical trial that evaluated the use of anastrozole for reduction of estrogen receptor–positive breast cancers in postmenopausal women at increased risk of developing breast cancer provided the predominant basis for the update. UPDATED RECOMMENDATIONS In postmenopausal women at increased risk, the choice of endocrine therapy now includes anastrozole (1 mg/day) in addition to exemestane (25 mg/day), raloxifene (60 mg/day), or tamoxifen (20 mg/day). The decision regarding choice of endocrine therapy should take into consideration age, baseline comorbidities, and adverse effect profiles. Clinicians should not prescribe anastrozole, exemestane, or raloxifene for breast cancer risk reduction to premenopausal women. Tamoxifen 20 mg/day for 5 years is still considered standard of care for risk reduction in premenopausal women who are at least 35 years old and have completed childbearing. Data on low-dose tamoxifen as an alternative to the standard dose for both pre- and postmenopausal women with intraepithelial neoplasia are discussed in the Clinical Considerations section of this article. Additional information is available at www.asco.org/breast-cancer-guidelines .
Collapse
Affiliation(s)
- Kala Visvanathan
- Johns Hopkins School of Medicine and Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | | | | | | | | | - Andrea DeCensi
- National Hospital E.O. Ospedali Galliera S.C. Oncologia Medica, Genoa, Italy; and Queen Mary University of London, United Kingdom
| | | | | | | | - Seema A. Khan
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | | | - Rachal Raab
- Cancer Care of Western North Carolina, Asheville, NC
| | | | | |
Collapse
|
292
|
Eraso Y. Oestrogen receptors and breast cancer: are we prepared to move forward? A critical review. BIOSOCIETIES 2019. [DOI: 10.1057/s41292-019-00173-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
293
|
Huang Y, Tong Z, Chen K, Wang Y, Liu P, Gu L, Liu J, Yu J, Song F, Zhao W, Shi Y, Li H, Xiao H, Hao X. Interpretation of breast cancer screening guideline for Chinese women. Cancer Biol Med 2019; 16:825-835. [PMID: 31908899 PMCID: PMC6936244 DOI: 10.20892/j.issn.2095-3941.2019.0322] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/26/2019] [Indexed: 12/19/2022] Open
Abstract
Breast cancer is the most common malignant tumor in Chinese women. Early screening is the best way to improve the rates of early diagnosis and survival of breast cancer patients. The peak onset age for breast cancer in Chinese women is considerably younger than those in European and American women. It is imperative to develop breast cancer screening guideline that is suitable for Chinese women. By summarizing the current evidence on breast cancer screening in Chinese women, and referring to the latest guidelines and consensus on breast cancer screening in Europe, the United States, and East Asia, the China Anti-Cancer Association and National Clinical Research Center for Cancer (Tianjin Medical University Cancer Institute and Hospital) have formulated population-based guideline for breast cancer screening in Chinese women. The guideline provides recommendations on breast cancer screening for Chinese women at average or high risk of breast cancer according to the following three aspects: age of screening, screening methods, and screening interval. This article provides more detailed information to support the recommendations in this guideline and to provide more direction for current breast cancer screening practices in China.
Collapse
Affiliation(s)
| | | | - Kexin Chen
- Department of Epidemiology and Statistics
| | - Ying Wang
- Department of Epidemiology and Statistics
- China Anti-Cancer Association, Tianjin 300060, China
| | | | - Lin Gu
- The 2 Surgery Department of Breast Oncology
| | | | - Jinpu Yu
- Cancer Molecular Diagnostics Core
| | | | - Wenhua Zhao
- Department of Epidemiology and Statistics
- China Anti-Cancer Association, Tianjin 300060, China
| | - Yehui Shi
- Medicine Department of Breast Oncology
| | - Hui Li
- Department of Gastrointestinal Cancer Biology
| | - Huaiyuan Xiao
- Department of Research and Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xishan Hao
- Department of Epidemiology and Statistics
- China Anti-Cancer Association, Tianjin 300060, China
| |
Collapse
|
294
|
Daly MB, Ross E. Breast Cancer Chemoprevention—Can We Make a Case for Precision Medicine? JAMA Oncol 2019; 5:1542-1544. [DOI: 10.1001/jamaoncol.2019.3785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Mary B. Daly
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Eric Ross
- Population Studies Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
295
|
Cocce KJ, Jasper JS, Desautels TK, Everett L, Wardell S, Westerling T, Baldi R, Wright TM, Tavares K, Yllanes A, Bae Y, Blitzer JT, Logsdon C, Rakiec DP, Ruddy DA, Jiang T, Broadwater G, Hyslop T, Hall A, Laine M, Phung L, Greene GL, Martin LA, Pancholi S, Dowsett M, Detre S, Marks JR, Crawford GE, Brown M, Norris JD, Chang CY, McDonnell DP. The Lineage Determining Factor GRHL2 Collaborates with FOXA1 to Establish a Targetable Pathway in Endocrine Therapy-Resistant Breast Cancer. Cell Rep 2019; 29:889-903.e10. [PMID: 31644911 PMCID: PMC6874102 DOI: 10.1016/j.celrep.2019.09.032] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 07/02/2019] [Accepted: 09/12/2019] [Indexed: 12/25/2022] Open
Abstract
Notwithstanding the positive clinical impact of endocrine therapies in estrogen receptor-alpha (ERα)-positive breast cancer, de novo and acquired resistance limits the therapeutic lifespan of existing drugs. Taking the position that resistance is nearly inevitable, we undertook a study to identify and exploit targetable vulnerabilities that were manifest in endocrine therapy-resistant disease. Using cellular and mouse models of endocrine therapy-sensitive and endocrine therapy-resistant breast cancer, together with contemporary discovery platforms, we identified a targetable pathway that is composed of the transcription factors FOXA1 and GRHL2, a coregulated target gene, the membrane receptor LYPD3, and the LYPD3 ligand, AGR2. Inhibition of the activity of this pathway using blocking antibodies directed against LYPD3 or AGR2 inhibits the growth of endocrine therapy-resistant tumors in mice, providing the rationale for near-term clinical development of humanized antibodies directed against these proteins.
Collapse
Affiliation(s)
- Kimberly J Cocce
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jeff S Jasper
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Taylor K Desautels
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Logan Everett
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Suzanne Wardell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Thomas Westerling
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Robert Baldi
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tricia M Wright
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kendall Tavares
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Alex Yllanes
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yeeun Bae
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | | | - Craig Logsdon
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Daniel P Rakiec
- Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA 02139, USA
| | - David A Ruddy
- Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA 02139, USA
| | - Tiancong Jiang
- Department of Biostatistics, Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gloria Broadwater
- Department of Biostatistics, Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Terry Hyslop
- Department of Biostatistics, Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Allison Hall
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Muriel Laine
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Linda Phung
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Geoffrey L Greene
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Lesley-Ann Martin
- Breast Cancer Now, Toby Robins Research Centre, Institute of Cancer Research, London, SW3 6JB, UK
| | - Sunil Pancholi
- Breast Cancer Now, Toby Robins Research Centre, Institute of Cancer Research, London, SW3 6JB, UK
| | - Mitch Dowsett
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital NHS Trust, London, SW3 6JJ, UK
| | - Simone Detre
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital NHS Trust, London, SW3 6JJ, UK
| | - Jeffrey R Marks
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gregory E Crawford
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Myles Brown
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - John D Norris
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
296
|
Abstract
Objective: This review describes historical development of selective estrogen receptor modulators (SERMs) and their combination with estrogens, termed a tissue selective estrogen complex (TSEC), and considers the potential for future TSEC development. Methods: This narrative review is based on literature identified on PubMed and the TSEC research and development experience of the authors. Results: SERMs have estrogenic and antiestrogenic effects in various tissues; however, no single agent has achieved an optimal balance of agonist and antagonist effects for the treatment of menopausal symptoms. Clinically, a number of SERMs protect against osteoporosis and breast cancer but can exacerbate vasomotor symptoms. Estrogens alleviate menopausal hot flushes and genitourinary symptoms as well as reduce bone loss, but the addition of a progestogen to menopausal hormone therapy to protect against endometrial cancer increases vaginal bleeding risk, breast tenderness, and potentially breast cancer. The search for an effective menopausal therapy with better tolerability led to the investigation of TSECs. Clinical development of a TSEC consisting of conjugated estrogens/bazedoxifene increased understanding of the importance of a careful consideration of the combination's components and their respective doses to balance safety and efficacy. Bazedoxifene is an estrogen receptor agonist in bone but an antagonist/degrader in the endometrium, which has contributed to its success as a TSEC component. Other oral TSEC combinations studied thus far have not demonstrated similar endometrial safety. Conclusions: Choice of SERM, selection of doses, and clinical trial data evaluating safety and efficacy are key to ensuring safety and adequate therapeutic effect of TSECs for addressing menopausal symptoms.
Collapse
|
297
|
Tingskov SJ, Mutsaers HAM, Nørregaard R. Estrogen regulates aquaporin-2 expression in the kidney. VITAMINS AND HORMONES 2019; 112:243-264. [PMID: 32061343 DOI: 10.1016/bs.vh.2019.08.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Estrogens are primarily identified as sex hormones that, for a long time, have been known as important regulators of female reproductive physiology. However, our understanding of the role of estrogens has changed over the past years. It is now well accepted that estrogens are also involved in other physiological and pathological processes in both genders. This is due to the fact that estrogen can act both local as well as on a systemic level. Next to its role in reproductive physiology, there is accumulating evidence that estrogen influences multiple systems involved in water homeostasis. This chapter will delineate the regulatory effects of estrogen on the water channel aquaporin-2 (AQP2) found in the renal collecting duct. We will first provide an introduction to estrogen, the estrogen receptors and their role in renal physiology as well as describe the effect of selective estrogen receptor modulators (SERMs) on the kidney. Subsequently, we will focus on how estrogen and SERMs influence water balance and regulate AQP2 expression in principal cells of the collecting duct. Finally, we will describe how estrogen regulates AQP2 functionality in other organ systems.
Collapse
Affiliation(s)
| | | | - Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
298
|
Mazzoni L, Giampieri F, Alvarez Suarez JM, Gasparrini M, Mezzetti B, Forbes Hernandez TY, Battino MA. Isolation of strawberry anthocyanin-rich fractions and their mechanisms of action against murine breast cancer cell lines. Food Funct 2019; 10:7103-7120. [PMID: 31621765 DOI: 10.1039/c9fo01721f] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The aim of this study was the evaluation of the effects of strawberry anthocyanin extract treatment on two in vitro models of murine breast cancer cell lines, in an attempt to detect a specific pathway (AMP-activated protein kinase or AMPK) through which strawberries exert their anticancer activity. The anticancer activity of purified anthocyanin extracts from an Alba cultivar on two murine cancer cell lines, N202/1A (with high levels of the HER2/neu oncogene) and N202/1E (with low levels of the HER2/neu oncogene), was evaluated after 48 and 72 h of treatment. The cell viability and apoptosis, intracellular ROS rates, and cell oxidative damage were assessed. Western blot assays were performed to analyze the expression of several proteins related to apoptosis, autophagy, metastasis, the oxidative status, mitochondrial functionality, and the AMPK pathway. This study demonstrated that the anthocyanin extract of Alba strawberry shows an antiproliferative effect on cancer cells, through the induction of apoptosis and oxidative stress, by stimulating different molecular pathways. This study is one of the first studies that have tried to deepen the understanding of a candidate pathway for the explanation of the effects of strawberry on cancer cells. A relationship between the AMPK pathway and the anticancer effects of strawberries was demonstrated.
Collapse
Affiliation(s)
- Luca Mazzoni
- Department of Agricultural, Food and Environmental Sciences - Università Politecnica delle Marche, Via Brecce Bianche 10, 60131, Ancona, Italy
| | - Francesca Giampieri
- Dipartimento di Scienze Cliniche Specialistiche e Odontostomatologiche - Università Politecnica delle Marche, Via Ruggeri, 60130, Ancona, Italy.
| | - Jose Miguel Alvarez Suarez
- Facultad de Ingeniería y Ciencias Aplicadas. Grupo de Investigación en Biotecnología Aplicada a Biomedicina, Universidad de Las Américas (UDLA), Quito, Ecuador
| | - Massimiliano Gasparrini
- Department of Agricultural, Food and Environmental Sciences - Università Politecnica delle Marche, Via Brecce Bianche 10, 60131, Ancona, Italy
| | - Bruno Mezzetti
- Department of Agricultural, Food and Environmental Sciences - Università Politecnica delle Marche, Via Brecce Bianche 10, 60131, Ancona, Italy
| | - Tamara Yuliett Forbes Hernandez
- Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo - Vigo Campus, 32004 Ourense, Spain.
| | - Maurizio Antonio Battino
- Dipartimento di Scienze Cliniche Specialistiche e Odontostomatologiche - Università Politecnica delle Marche, Via Ruggeri, 60130, Ancona, Italy. and Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo - Vigo Campus, 32004 Ourense, Spain. and College of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
299
|
Chollet J, Mermelstein F, Rocamboli SC, Friend DR. Vaginal tamoxifen for treatment of vulvar and vaginal atrophy: Pharmacokinetics and local tolerance in a rabbit model over 28 days. Int J Pharm 2019; 570:118691. [DOI: 10.1016/j.ijpharm.2019.118691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/03/2019] [Accepted: 09/08/2019] [Indexed: 11/15/2022]
|
300
|
Gail MH, Pfeiffer RM. Breast Cancer Risk Model Requirements for Counseling, Prevention, and Screening. J Natl Cancer Inst 2019; 110:994-1002. [PMID: 29490057 DOI: 10.1093/jnci/djy013] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/16/2018] [Indexed: 01/05/2023] Open
Abstract
Background Incorporation of polygenic risk scores and mammographic density into models to predict breast cancer incidence can increase discriminatory accuracy (area under the receiver operating characteristic curve [AUC]) from 0.6 for models based only on epidemiologic factors to 0.7. It is timely to assess what impact these improvements will have on individual counseling and on public health prevention and screening strategies, and to determine what further improvements are needed. Methods We studied various clinical and public health applications using a log-normal distribution of risk. Results Provided they are well calibrated, even risk models with AUCs of 0.6 to 0.7 provide useful perspective for individual counseling and for weighing the harms and benefits of preventive interventions in the clinic. At the population level, they are helpful for designing preventive intervention trials, for assessing reductions in absolute risk from reducing exposure to modifiable risk factors, and for resource allocation (although a higher AUC would be desirable for risk-based allocation). Other public health applications require higher AUCs that can only be achieved with risk predictors 1.6 to 8.8 times as strong as all those yet discovered combined. Such applications are preventing an appreciable proportion of population disease when employing a high-risk prevention strategy and deciding who should be screened for subclinical disease. Conclusions Current and foreseeable risk models are useful for counseling and some prevention activities, but given the daunting challenge of achieving, for example, an AUC of 0.8, considerable effort should be put into finding effective preventive interventions and screening strategies with fewer adverse effects.
Collapse
Affiliation(s)
- Mitchell H Gail
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | - Ruth M Pfeiffer
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| |
Collapse
|