1
|
Tomida S, Koyama T, Ozaki E, Takashima N, Morita M, Sakaguchi K, Naoi Y, Nishida Y, Hara M, Hishida A, Tamura T, Okada R, Kubo Y, Otonari J, Ikezaki H, Nakamura Y, Kusakabe M, Tanoue S, Koriyama C, Koyanagi YN, Ito H, Suzuki S, Otani T, Miyagawa N, Okami Y, Arisawa K, Watanabe T, Kuriki K, Wakai K, Matsuo K. Seven-plus hours of daily sedentary time and the subsequent risk of breast cancer: Japan Multi-Institutional Collaborative Cohort Study. Cancer Sci 2024; 115:611-622. [PMID: 38041484 PMCID: PMC10859602 DOI: 10.1111/cas.16020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/15/2023] [Accepted: 11/06/2023] [Indexed: 12/03/2023] Open
Abstract
This study aimed to investigate the association between daily sedentary time and the risk of breast cancer (BC) in a large Japanese population. The participants were 36,023 women aged 35-69 years from the Japan Multi-Institutional Collaborative Cohort Study. Cox proportional hazards analysis was used to estimate adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) for BC incidence in relation to time spent sedentarily (categorical variables: <7 and ≥7 hours/day [h/d]). Additionally, the associations of BC incidence to the joint effect of sedentary time with each component of physical activity, such as leisure-time metabolic equivalents (METs), frequency of leisure-time physical activity, and daily walking time, were examined. During 315,189 person-years of follow-up, 554 incident cases of BC were identified. When compared to participants who spent <7 h/d sedentary, those who spent ≥7 h/d sedentary have a significantly higher risk of BC (HR, 1.36; 95% CI, 1.07-1.71). The corresponding HRs among participants who spent ≥7 h/d sedentary with more physical activity, such as ≥1 h/d for leisure-time METs, ≥3 days/week of leisure-time physical activity, and ≥1 h/d of daily walking were 1.58 (95% CI, 1.11-2.25), 1.77 (95% CI, 1.20-2.61), and 1.42 (95% CI, 1.10-1.83), respectively, compared with those who spent <7 h/d sedentary. This study found that spending ≥7 h/d of sedentary time is associated with the risk of BC. Neither leisure-time physical activity nor walking had a BC-preventive effect in those with ≥7 h/d of sedentary time.
Collapse
Affiliation(s)
- Satomi Tomida
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Teruhide Koyama
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Etsuko Ozaki
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Naoyuki Takashima
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Public Health, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Midori Morita
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Koichi Sakaguchi
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuto Naoi
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuichiro Nishida
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Megumi Hara
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Asahi Hishida
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Tamura
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rieko Okada
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoko Kubo
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jun Otonari
- Department of Psychosomatic Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroaki Ikezaki
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
- Department of Comprehensive General Internal Medicine, Kyushu University Faculty of Medical Sciences, Fukuoka, Japan
| | - Yohko Nakamura
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Miho Kusakabe
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Shiroh Tanoue
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Chihaya Koriyama
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yuriko N Koyanagi
- Division of Cancer Information and Control, Aichi Cancer Center Research Institute, Nagoya, Aichi, Japan
| | - Hidemi Ito
- Division of Cancer Information and Control, Aichi Cancer Center Research Institute, Nagoya, Aichi, Japan
- Division of Descriptive Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Sadao Suzuki
- Department of Public Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Takahiro Otani
- Department of Public Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Naoko Miyagawa
- Department of Public Health, Shiga University of Medical Science, Otsu, Shiga, Japan
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Yukiko Okami
- NCD Epidemiology Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Kokichi Arisawa
- Department of Preventive Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takeshi Watanabe
- Department of Preventive Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kiyonori Kuriki
- Laboratory of Public Health, Division of Nutritional Sciences, School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kenji Wakai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center, Nagoya, Aichi, Japan
- Department of Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|
2
|
Akingbesote ND, Owusu D, Liu R, Cartmel B, Ferrucci LM, Zupa M, Lustberg MB, Sanft T, Blenman KRM, Irwin ML, Perry RJ. A review of the impact of energy balance on triple-negative breast cancer. J Natl Cancer Inst Monogr 2023; 2023:104-124. [PMID: 37139977 DOI: 10.1093/jncimonographs/lgad011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 05/05/2023] Open
Abstract
Cancer cells cannot proliferate without sufficient energy to generate biomass for rapid cell division, as well as to fuel their functions at baseline. For this reason, many recent observational and interventional studies have focused on increasing energy expenditure and/or reducing energy intake during and after cancer treatment. The impact of variance in diet composition and in exercise on cancer outcomes has been detailed extensively elsewhere and is not the primary focus of this review. Instead, in this translational, narrative review we examine studies of how energy balance impacts anticancer immune activation and outcomes in triple-negative breast cancer (TNBC). We discuss preclinical, clinical observational, and the few clinical interventional studies on energy balance in TNBC. We advocate for the implementation of clinical studies to examine how optimizing energy balance-through changes in diet and/or exercise-may optimize the response to immunotherapy in people with TNBC. It is our conviction that by taking a holistic approach that includes energy balance as a key factor to be considered during and after treatment, cancer care may be optimized, and the detrimental effects of cancer treatment and recovery on overall health may be minimized.
Collapse
Affiliation(s)
- Ngozi D Akingbesote
- Department of Internal Medicine, Yale University, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, USA
| | - Dennis Owusu
- Department of Internal Medicine, Yale University, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, USA
- Kwame Nkrumah University of Science and Technology, Kumasi, Ashanti Region, Ghana
| | - Ryan Liu
- Department of Internal Medicine, Yale University, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, USA
- Cedar Park High School, Cedar Park, TX, USA
| | - Brenda Cartmel
- Yale School of Public Health, New Haven, CT, USA
- Yale Cancer Center, New Haven, CT, USA
| | - Leah M Ferrucci
- Yale School of Public Health, New Haven, CT, USA
- Yale Cancer Center, New Haven, CT, USA
| | | | - Maryam B Lustberg
- Department of Internal Medicine, Yale University, New Haven, CT, USA
- Yale Cancer Center, New Haven, CT, USA
| | - Tara Sanft
- Department of Internal Medicine, Yale University, New Haven, CT, USA
- Yale Cancer Center, New Haven, CT, USA
| | - Kim R M Blenman
- Department of Internal Medicine, Yale University, New Haven, CT, USA
- Yale Cancer Center, New Haven, CT, USA
- Department of Computer Science, Yale University, New Haven, CT, USA
| | - Melinda L Irwin
- Yale School of Public Health, New Haven, CT, USA
- Yale Cancer Center, New Haven, CT, USA
| | - Rachel J Perry
- Department of Internal Medicine, Yale University, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, USA
- Yale Cancer Center, New Haven, CT, USA
| |
Collapse
|
3
|
Obr AE, Bulatowicz JJ, Chang YJ, Ciliento V, Lemenze A, Maingrette K, Shang Q, Gallagher EJ, LeRoith D, Wood TL. Breast tumor IGF1R regulates cell adhesion and metastasis: alignment of mouse single cell and human breast cancer transcriptomics. Front Oncol 2022; 12:990398. [PMID: 36568144 PMCID: PMC9769962 DOI: 10.3389/fonc.2022.990398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction The acquisition of a metastatic phenotype is the critical event that determines patient survival from breast cancer. Several receptor tyrosine kinases have functions both in promoting and inhibiting metastasis in breast tumors. Although the insulin-like growth factor 1 receptor (IGF1R) has been considered a target for inhibition in breast cancer, low levels of IGF1R expression are associated with worse overall patient survival. Methods To determine how reduced IGF1R impacts tumor phenotype in human breast cancers, we used weighted gene co-expression network analysis (WGCNA) of Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) patient data to identify gene modules associated with low IGF1R expression. We then compared these modules to single cell gene expression analyses and phenotypes of mouse mammary tumors with reduced IGF1R signaling or expression in a tumor model of triple negative breast cancer. Results WGCNA from METABRIC data revealed gene modules specific to cell cycle, adhesion, and immune cell signaling that were inversely correlated with IGF1R expression in human breast cancers. Integration of human patient data with single cell sequencing data from mouse tumors revealed similar pathways necessary for promoting metastasis in basal-like mammary tumors with reduced signaling or expression of IGF1R. Functional analyses revealed the basis for the enhanced metastatic phenotype including alterations in E- and P-cadherins. Discussion Human breast and mouse mammary tumors with reduced IGF1R are associated with upregulation of several pathways necessary for promoting metastasis supporting the conclusion that IGF1R normally helps maintain a metastasis suppressive tumor microenvironment. We further found that reduced IGF1R signaling in tumor epithelial cells dysregulates cadherin expression resulting in reduced cell adhesion.
Collapse
Affiliation(s)
- Alison E. Obr
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Joseph J. Bulatowicz
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Yun-Juan Chang
- Office of Advance Research Computing, Rutgers University, Piscataway, NJ, United States
| | - Virginia Ciliento
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Alexander Lemenze
- Department of Pathology, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Krystopher Maingrette
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Quan Shang
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Emily J. Gallagher
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Icahn Sinai School of Medicine at Mt. Sinai, New York, NY, United States
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Icahn Sinai School of Medicine at Mt. Sinai, New York, NY, United States
| | - Teresa L. Wood
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, United States,*Correspondence: Teresa L. Wood,
| |
Collapse
|
4
|
Ennis CS, Llevenes P, Qiu Y, Dries R, Denis GV. The crosstalk within the breast tumor microenvironment in type II diabetes: Implications for cancer disparities. Front Endocrinol (Lausanne) 2022; 13:1044670. [PMID: 36531496 PMCID: PMC9751481 DOI: 10.3389/fendo.2022.1044670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/17/2022] [Indexed: 12/04/2022] Open
Abstract
Obesity-driven (type 2) diabetes (T2D), the most common metabolic disorder, both increases the incidence of all molecular subtypes of breast cancer and decreases survival in postmenopausal women. Despite this clear link, T2D and the associated dysfunction of diverse tissues is often not considered during the standard of care practices in oncology and, moreover, is treated as exclusion criteria for many emerging clinical trials. These guidelines have caused the biological mechanisms that associate T2D and breast cancer to be understudied. Recently, it has been illustrated that the breast tumor microenvironment (TME) composition and architecture, specifically the surrounding cellular and extracellular structures, dictate tumor progression and are directly relevant for clinical outcomes. In addition to the epithelial cancer cell fraction, the breast TME is predominantly made up of cancer-associated fibroblasts, adipocytes, and is often infiltrated by immune cells. During T2D, signal transduction among these cell types is aberrant, resulting in a dysfunctional breast TME that communicates with nearby cancer cells to promote oncogenic processes, cancer stem-like cell formation, pro-metastatic behavior and increase the risk of recurrence. As these cells are non-malignant, despite their signaling abnormalities, data concerning their function is never captured in DNA mutational databases, thus we have limited insight into mechanism from publicly available datasets. We suggest that abnormal adipocyte and immune cell exhaustion within the breast TME in patients with obesity and metabolic disease may elicit greater transcriptional plasticity and cellular heterogeneity within the expanding population of malignant epithelial cells, compared to the breast TME of a non-obese, metabolically normal patient. These challenges are particularly relevant to cancer disparities settings where the fraction of patients seen within the breast medical oncology practice also present with co-morbid obesity and metabolic disease. Within this review, we characterize the changes to the breast TME during T2D and raise urgent molecular, cellular and translational questions that warrant further study, considering the growing prevalence of T2D worldwide.
Collapse
Affiliation(s)
- Christina S. Ennis
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
- Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, United States
| | - Pablo Llevenes
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Yuhan Qiu
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
| | - Ruben Dries
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
- Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, United States
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA, United States
| | - Gerald V. Denis
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Shipley Prostate Cancer Research Professor, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
5
|
Yang PJ, Hou MF, Ou-Yang F, Tsai EM, Wang TN. Association of early-onset breast cancer with body mass index, menarche, and menopause in Taiwan. BMC Cancer 2022; 22:259. [PMID: 35277131 PMCID: PMC8917681 DOI: 10.1186/s12885-022-09361-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 02/28/2022] [Indexed: 12/20/2022] Open
Abstract
Background The trend of women suffering from early-onset breast cancer is increasing in Taiwan. The association of early-onset breast cancer with body mass index (BMI), menarche, and menopausal status has focused interest on the field of cancer epidemiology; however, few studies have explored the interaction of these factors on early-onset risk. This study aimed to estimate the interaction effects of BMI, menarche, and menopausal status on 40-year-old early-onset breast cancer. Methods Breast cancer patients were recruited from Kaohsiung Medical University Chung-Ho Memorial Hospital from 2013 to 2020. Multivariable logistic regression was used to estimate odds ratios (ORs) for early-onset breast cancer risk associated with menarcheal age stratified by sociodemographic factors and for the interaction between BMI and menopausal status on early-onset risk. Results A total of 775 participants were divided into 131 early-onset cases (≤ 40 years) and 644 late-onset cases (> 40 years). Compared to the age of 13 years at menarche, the age ≤ 11 years was significantly positively associated (OR: 2.62, 95% CI: 1.38–4.97) and ≥ 16 years was negatively associated (OR: 0.13, 95% CI: 0.03–0.53) with 40-year-old early-onset breast cancer respectively. In an adjusted model, the status of BMI < 24 and premenopause had 1.76- and 4.59-fold risk of early-onset breast cancer respectively. Especially in BMI < 24 status, premenopause also had a 6.47-fold early-onset risk and the early-onset risk increased by a significant amount per one year younger at menarche (aOR: 1.26, 95% CI: 1.03–1.55). There was also a positive interaction effect on an additive scale between BMI and menopausal status on early-onset breast cancer (RERIOR = 4.62, Pinteraction = 0.057). Compared to both BMI ≥ 24 and peri-/postmenopausal status, both the status of BMI < 24 and premenopause were associated with early-onset breast cancer (aOR: 7.16, 95% CI: 3.87–13.25). Conclusions This study suggests that the status of BMI < 24 and premenopause were associated with an increased risk of early-onset breast cancer and there was a positive interaction on an additive scale. Understanding how obesity and menopausal status affect early-onset breast cancer is important for drafting preventive measures for early-onset breast cancer in Taiwan.
Collapse
|
6
|
Abstract
The breast and ovarian cancer susceptibility gene (BRCA1) is a tumor suppressor whose mutation has been associated with the development of breast, ovarian and, probably, other malignancies at young ages. The BRCA1 gene product participates in multiple biological pathways including the DNA damage response, transcriptional control, cell growth and apoptosis. Inactivating germline mutations of the BRCA1 gene can be detected in a substantial portion of families with inherited breast and/or ovarian cancer. While the genomic and cancer-related actions of BRCA1 have been extensively investigated, not much information exists regarding the cellular and circulating factors involved in regulation of BRCA1 expression and action. The present review article dissects the emerging role of BRCA1 as an important regulator of various endocrine and metabolic axes. Experimental and clinical evidence links BRCA1 with a number of peptide and steroid hormones. Furthermore, comprehensive analyses identified complex interactions between the insulin/insulin-like growth factor-1 (IGF1) signaling axis and BRCA1. The correlation between metabolic disorders, including diabetes and the metabolic syndrome, and BRCA1 mutations, are discussed in this article.
Collapse
|
7
|
Ianza A, Sirico M, Bernocchi O, Generali D. Role of the IGF-1 Axis in Overcoming Resistance in Breast Cancer. Front Cell Dev Biol 2021; 9:641449. [PMID: 33829018 PMCID: PMC8019779 DOI: 10.3389/fcell.2021.641449] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/04/2021] [Indexed: 12/12/2022] Open
Abstract
Over the last two decades, many studies have demonstrated that the insulin-like growth factor-1 (IGF-1) is involved in a number of patho-physiological processes, as well as in the development of different types of solid tumors, including breast cancer (BC). Preclinical and clinical data showed that IGF-1 receptor (R) is overexpressed and hyper-phosphorylated in several subtypes of BCs. The central implications of this pathway in tumor cell proliferation and metastasis make it an important therapeutic target. Moreover, the IGF-1 axis has shown strong interconnection with estrogen regulation and endocrine therapy, suggesting a possible solution to anti-estrogen resistance. IGF-1R might also interfere with other pivotal therapeutic strategies, such as anti HER2 treatments and mTOR inhibitors; several clinical trials are ongoing evaluating the role of IGF-1R inhibition in modulating resistance mechanisms to target therapies. Our aim is to offer an overview of the most recent and significant field of application of IGF-1 inhibitors and relevant therapeutic strategies, weighing their possible future impact on clinical practice.
Collapse
Affiliation(s)
- Anna Ianza
- Department of Medical, Surgery and Health Sciences, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Marianna Sirico
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
- Breast Cancer Unit and Translational Research Unit, ASST Cremona, Cremona, Italy
| | - Ottavia Bernocchi
- Department of Medical, Surgery and Health Sciences, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Daniele Generali
- Department of Medical, Surgery and Health Sciences, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
- Breast Cancer Unit and Translational Research Unit, ASST Cremona, Cremona, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
8
|
Abdullah AZ, Fitri SA, Muniroh M, Agustini TW. Patin (Pangasius hypophthalmus) fish protein concentrate alters insulin-like growth factor (IGF)-1 and igf binding protein (IGFBP)-3 level of sprague dawley neonate rats-induced malnutrition. POTRAVINARSTVO 2020. [DOI: 10.5219/1394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Malnutrition is caused by inadequate protein intake and affects growth factor. High protein from patin (Pangasius hypophthalmus) fish is a well-known protein source. This study aims to investigate the effect of patin fish protein concentrate (PFPC) in the IGF-1 and IGFBP-3 level of Sprague Dawley (SD) neonate rats-induced malnutrition. Thirty male SD neonate rats were divided randomly into five groups, namely normal control (K1), malnutrition control (K2), malnutrition with PFPC 13.26 mg.g-1 body weight (BW)/day (X1), malnutrition with PFPC 19.89 mg.g-1 BW/d (X2), and malnutrition with casein supplement 13.26 mg.g-1 BW/d (X3). K1 received a standard diet, while the others received a low 8% protein diet (L8PD) since those were born until 21 days. The standard diet was refed for all groups during the intervention (14 days). IGF-1 and IGFBP-3 levels were measured by ELISA. Normal data were analyzed by using One-way ANOVA which then was followed by post-hoc Bonferroni. Meanwhile, the others were analyzed by Kruskal Wallis and followed by Mann-Whitney U-test. Spearman test was used for correlation. PFPC contained 81.07% of protein, 4.08% of fat, 7.24% of moisture, 2.77% of ash, and 4.83% of carbohydrate. These contents had affected the growth factor. As a result, in the PFPC intervention, IGF-1, and IGFBP-3 levels (p <0.05) were decreased, while the controls were increased. The decreased values were shown in IGFBP-3 levels (p <0.05) while the increase was shown in both controls. On the other hand, the increase in body weight was shown in all groups, including control ones. A strong correlation was found between IGF-1 and IGFBP-3. PFPC has additional value on repairing malnutrition that is the best dose in effecting IGF-1 dan IGFBP3 levels is 13.26 mg.g-1 BW/d.
Collapse
|
9
|
Harguindey S, Alfarouk K, Polo Orozco J, Fais S, Devesa J. Towards an Integral Therapeutic Protocol for Breast Cancer Based upon the New H +-Centered Anticancer Paradigm of the Late Post-Warburg Era. Int J Mol Sci 2020; 21:E7475. [PMID: 33050492 PMCID: PMC7589677 DOI: 10.3390/ijms21207475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
A brand new approach to the understanding of breast cancer (BC) is urgently needed. In this contribution, the etiology, pathogenesis, and treatment of this disease is approached from the new pH-centric anticancer paradigm. Only this unitarian perspective, based upon the hydrogen ion (H+) dynamics of cancer, allows for the understanding and integration of the many dualisms, confusions, and paradoxes of the disease. The new H+-related, wide-ranging model can embrace, from a unique perspective, the many aspects of the disease and, at the same time, therapeutically interfere with most, if not all, of the hallmarks of cancer known to date. The pH-related armamentarium available for the treatment of BC reviewed here may be beneficial for all types and stages of the disease. In this vein, we have attempted a megasynthesis of traditional and new knowledge in the different areas of breast cancer research and treatment based upon the wide-ranging approach afforded by the hydrogen ion dynamics of cancer. The concerted utilization of the pH-related drugs that are available nowadays for the treatment of breast cancer is advanced.
Collapse
Affiliation(s)
- Salvador Harguindey
- Department of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
| | - Khalid Alfarouk
- Department of Pharmacology, Al-Ghad International Colleges for Applied Medical Sciences, Al-Madinah Al-Munawarah 42316, Saudi Arabia and Alfarouk Biomedical Research LLC, Tampa, FL 33617, USA;
| | - Julián Polo Orozco
- Department of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (National Institute of Health), 00161 Rome, Italy;
| | - Jesús Devesa
- Scientific Direction, Foltra Medical Centre, 15886 Teo, Spain;
| |
Collapse
|
10
|
The effect of synbiotic on glycemic profile and sex hormones in overweight and obese breast cancer survivors following a weight-loss diet: A randomized, triple-blind, controlled trial. Clin Nutr 2020; 40:394-403. [PMID: 32698957 DOI: 10.1016/j.clnu.2020.05.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The investigation was designed to assess the effects of synbiotic supplementation on glycemic profile, insulin-like growth factor-1 (IGF-1) and sex hormones in overweight and obese postmenopausal breast cancer survivors (BCSs) who had hormone-receptor-positive breast cancer. METHODS This randomized, triple-blind, placebo-controlled trial was conducted on 76 overweight and obese BCSs aged 57.43 (5.82) years. All participants were given a specified low calorie diet and were randomly assigned into two groups to intake 109 CFU/day of synbiotic supplement (n = 38) or placebo (n = 38) for 8 weeks. Body composition, physical activity, glycemic profile, IGF-1, estradiol, testosterone and dehydroepiandrosterone sulfate (DHEA-S) were measured at baseline and after 8 weeks. RESULTS A significant reduction in serum insulin (median change (Q1, Q3) from baseline of -1.05 (-2.36, 0.32) μIU/mL; P = 0.006) and insulin resistance (HOMA-IR) (mean change (SD) from baseline of -4.0 (0.9); P = 0.007) were seen over the 8 weeks in the synbiotic group. However, no significant changes were observed in serum insulin, fasting plasma glucose, HbA1c, HOMA-IR, IGF-1, estradiol, testosterone, DHEA-S and sex hormone binding globulin between-groups at the end of the intervention. CONCLUSIONS Overall, as the 8-week synbiotic consumption compared with placebo had insignificant-reducing effects on glycemic profile, IGF-1 and sex hormones among overweight and obese postmenopausal BCSs, synbiotics may exert considerable beneficial consequences, which need to be further assessed in future clinical trials. TRIAL REGISTRATION IRCT, IRCT2015090223861N1. Registered 02 February 2017, http://www.irct.ir: IRCT2015090223861N1.
Collapse
|
11
|
Farvid MS, Spence ND, Holmes MD, Barnett JB. Fiber consumption and breast cancer incidence: A systematic review and meta-analysis of prospective studies. Cancer 2020; 126:3061-3075. [PMID: 32249416 DOI: 10.1002/cncr.32816] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/12/2020] [Accepted: 01/14/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Associations between fiber intake and breast cancer risk have been evaluated in prospective studies, but overall, the evidence is inconsistent. The authors performed a systematic review and meta-analysis of prospective studies to investigate the relation between intake of total and types of fiber with breast cancer incidence. METHODS The MEDLINE and Excerpta Medica dataBASE (EMBASE) databases were searched through July 2019 for prospective studies that reported on the association between fiber consumption and incident breast cancer. The pooled relative risk (RR) and 95% confidence intervals (95% CI) were estimated comparing the highest versus the lowest category of total and types of fiber consumption, using a random-effects meta-analysis. RESULTS The authors identified 17 cohort studies, 2 nested case-control studies, and 1 clinical trial study. Total fiber consumption was associated with an 8% lower risk of breast cancer (comparing the highest versus the lowest category, pooled RR, 0.92; 95% CI, 0.88-0.95 [I2 = 12.6%]). Soluble fiber was found to be significantly inversely associated with risk of breast cancer (pooled RR, 0.90 [95% CI, 0.84-0.96; I2 = 12.6%]) and insoluble fiber was found to be suggestively inversely associated with risk of breast cancer (pooled RR, 0.93 [95% CI, 0.86-1.00; I2 = 33.4%]). Higher total fiber intake was associated with a lower risk of both premenopausal and postmenopausal breast cancers (pooled RR, 0.82 [95% CI, 0.67-0.99; I2 = 35.2%] and pooled RR, 0.91 [95% CI, 0.88-0.95; I2 = 0.0%], respectively). Furthermore, the authors observed a nonsignificant inverse association between intake of total fiber and risk of both estrogen and progesterone receptor-positive and estrogen and progesterone receptor-negative breast cancers. CONCLUSIONS A random-effects meta-analysis of prospective observational studies demonstrated that high total fiber consumption was associated with a reduced risk of breast cancer. This finding was consistent for soluble fiber as well as for women with premenopausal and postmenopausal breast cancer.
Collapse
Affiliation(s)
- Maryam S Farvid
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Nicholas D Spence
- Department of Sociology and Interdisciplinary Center for Health and Society, University of Toronto, Toronto, Ontario, Canada
| | - Michelle D Holmes
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Junaidah B Barnett
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| |
Collapse
|
12
|
Nieuwenhuis S, Okkersen K, Widomska J, Blom P, 't Hoen PAC, van Engelen B, Glennon JC. Insulin Signaling as a Key Moderator in Myotonic Dystrophy Type 1. Front Neurol 2019; 10:1229. [PMID: 31849810 PMCID: PMC6901991 DOI: 10.3389/fneur.2019.01229] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/05/2019] [Indexed: 12/15/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is an autosomal dominant genetic disease characterized by multi-system involvement. Affected organ system includes skeletal muscle, heart, gastro-intestinal system and the brain. In this review, we evaluate the evidence for alterations in insulin signaling and their relation to clinical DM1 features. We start by summarizing the molecular pathophysiology of DM1. Next, an overview of normal insulin signaling physiology is given, and evidence for alterations herein in DM1 is presented. Clinically, evidence for involvement of insulin signaling pathways in DM1 is based on the increased incidence of insulin resistance seen in clinical practice and recent trial evidence of beneficial effects of metformin on muscle function. Indirectly, further support may be derived from certain CNS derived symptoms characteristic of DM1, such as obsessive-compulsive behavior features, for which links with altered insulin signaling has been demonstrated in other diseases. At the basic scientific level, several pathophysiological mechanisms that operate in DM1 may compromise normal insulin signaling physiology. The evidence presented here reflects the importance of insulin signaling in relation to clinical features of DM1 and justifies further basic scientific and clinical, therapeutically oriented research.
Collapse
Affiliation(s)
- Sylvia Nieuwenhuis
- Department of Cognitive Neuroscience, Donders Institute for Brain Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Kees Okkersen
- Department of Neurology, Donders Institute for Brain Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Joanna Widomska
- Department of Cognitive Neuroscience, Donders Institute for Brain Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Paul Blom
- VDL Enabling Technologies Group B.V., Eindhoven, Netherlands
| | - Peter A C 't Hoen
- Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Baziel van Engelen
- Department of Neurology, Donders Institute for Brain Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Jeffrey C Glennon
- Department of Cognitive Neuroscience, Donders Institute for Brain Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| |
Collapse
|
13
|
Farvid MS, Eliassen AH, Cho E, Chen WY, Willett WC. Dairy Consumption in Adolescence and Early Adulthood and Risk of Breast Cancer. Cancer Epidemiol Biomarkers Prev 2019; 27:575-584. [PMID: 29716928 DOI: 10.1158/1055-9965.epi-17-0345] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/15/2017] [Accepted: 03/05/2018] [Indexed: 01/07/2023] Open
Abstract
Background: Carcinogenic exposure in early life may be critical for subsequent breast cancer risk. Dairy consumption was examined during adolescence and early adulthood in relation to incident breast cancer in the Nurses' Health Study II cohort.Methods: For the analyses of early adulthood dairy consumption, we included 90,503 premenopausal women ages 27 to 44 years in 1991 who reported dairy consumption using a validated food-frequency questionnaire. From 1991 to 2013, 3,191 invasive breast cancer cases were identified. In 1998, 44,264 women recalled adolescent dairy consumption. This subgroup of women was followed up from 1998 to 2013; 1,318 invasive breast cancer cases were identified. Multivariate hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated using the Cox proportional hazard regression.Results: Adolescent and early adulthood total dairy consumption was not associated with overall breast cancer risk (each serving/day during adolescence, total dairy HR = 1.02, 95% CI, 0.97-1.07; for early adulthood total dairy HR = 1.01, 95% CI, 0.97-1.04), as were intakes of calcium, vitamin D, and lactose. Adolescent consumption of total and high-fat dairy was associated with higher risk of estrogen and progesterone receptor negative (each serving/day: total dairy HR = 1.11, 95% CI, 1.00-1.24; high-fat dairy HR = 1.17, 95% CI, 1.04-1.31). However, higher adolescent high-fat dairy consumption was associated with lower risk of estrogen and progesterone receptor positive tumors (each serving/day HR = 0.91, 95% CI, 0.86-0.97).Conclusions: Our results suggest no overall association between dairy consumption during adolescence or early adulthood and breast cancer risk, but the findings may differ by hormone receptor status of tumors.Impact: Dairy consumption in adolescence or early adulthood may not be a significant predictor of breast cancer incidence. Cancer Epidemiol Biomarkers Prev; 27(5); 575-84. ©2018 AACR.
Collapse
Affiliation(s)
- Maryam S Farvid
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.
| | - A Heather Eliassen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Eunyoung Cho
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Dermatology, The Warren Alpert Medical School of Brown University, Providence, Rhode Island.,Department of Epidemiology, Brown University School of Public Health, Providence, Rhode Island
| | - Wendy Y Chen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Walter C Willett
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
14
|
Obr AE, Kumar S, Chang YJ, Bulatowicz JJ, Barnes BJ, Birge RB, Lazzarino DA, Gallagher E, LeRoith D, Wood TL. Insulin-like growth factor receptor signaling in breast tumor epithelium protects cells from endoplasmic reticulum stress and regulates the tumor microenvironment. Breast Cancer Res 2018; 20:138. [PMID: 30458886 PMCID: PMC6245538 DOI: 10.1186/s13058-018-1063-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/18/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Early analyses of human breast cancer identified high expression of the insulin-like growth factor type 1 receptor (IGF-1R) correlated with hormone receptor positive breast cancer and associated with a favorable prognosis, whereas low expression of IGF-1R correlated with triple negative breast cancer (TNBC). We previously demonstrated that the IGF-1R acts as a tumor and metastasis suppressor in the Wnt1 mouse model of TNBC. The mechanisms for how reduced IGF-1R contributes to TNBC phenotypes is unknown. METHODS We analyzed the METABRIC dataset to further stratify IGF-1R expression with patient survival and specific parameters of TNBC. To investigate molecular events associated with the loss of IGF-1R function in breast tumor cells, we inhibited IGF-1R in human cell lines using an IGF-1R blocking antibody and analyzed MMTV-Wnt1-mediated mouse tumors with reduced IGF-1R function through expression of a dominant-negative transgene. RESULTS Our analysis of the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset revealed association between low IGF-1R and reduced overall patient survival. IGF-1R expression was inversely correlated with patient survival even within hormone receptor-positive breast cancers, indicating reduced overall patient survival with low IGF-1R was not due simply to low IGF-1R expression within TNBCs. Inhibiting IGF-1R in either mouse or human tumor epithelial cells increased reactive oxygen species (ROS) production and activation of the endoplasmic reticulum stress response. IGF-1R inhibition in tumor epithelial cells elevated interleukin (IL)-6 and C-C motif chemokine ligand 2 (CCL2) expression, which was reversed by ROS scavenging. Moreover, the Wnt1/dnIGF-1R primary tumors displayed a tumor-promoting immune phenotype. The increased CCL2 promoted an influx of CD11b+ monocytes into the primary tumor that also had increased matrix metalloproteinase (MMP)-2, MMP-3, and MMP-9 expression. Increased MMP activity in the tumor stroma was associated with enhanced matrix remodeling and collagen deposition. Further analysis of the METABRIC dataset revealed an increase in IL-6, CCL2, and MMP-9 expression in patients with low IGF-1R, consistent with our mouse tumor model and data in human breast cancer cell lines. CONCLUSIONS Our data support the hypothesis that reduction of IGF-1R function increases cellular stress and cytokine production to promote an aggressive tumor microenvironment through infiltration of immune cells and matrix remodeling.
Collapse
Affiliation(s)
- Alison E Obr
- Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, 07101, USA
| | - Sushil Kumar
- Department of Microbiology, Biochemistry & Molecular Genetics, Rutgers-New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, 07101, USA
| | - Yun-Juan Chang
- Office of Advance Research Computing, Rutgers-New Jersey Medical School, Newark, NJ, 07102, USA
| | - Joseph J Bulatowicz
- Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, 07101, USA
| | - Betsy J Barnes
- Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Raymond B Birge
- Department of Microbiology, Biochemistry & Molecular Genetics, Rutgers-New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, 07101, USA
| | - Deborah A Lazzarino
- Department of Microbiology, Biochemistry & Molecular Genetics, Rutgers-New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, 07101, USA
| | - Emily Gallagher
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Icahn Sinai School of Medicine at Mt. Sinai, New York, NY, 10029, USA
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Icahn Sinai School of Medicine at Mt. Sinai, New York, NY, 10029, USA
| | - Teresa L Wood
- Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, 07101, USA.
| |
Collapse
|
15
|
Abstract
The important role of insulin-like growth factor 1 receptor (IGF-1R) in malignant tumors has been well established. Increased IGF-1R activity promotes cancer cell proliferation, migration, and invasion and is associated with tumor metastasis, treatment resistance, poor prognosis, and shortened survival in patients with cancer. However, while IGF-1R has become a promising target for cancer therapy, IGF-1R-targeted therapy is ineffective in unselected patients. It is therefore essential to evaluate IGF-1R expression before treatment in order to identify responsive patients, monitor therapy efficacy, and estimate prognosis. Insulin-like growth factor 1 receptor molecular imaging is an optimal method for assessing the expression of IGF-1R in vivo accurately and noninvasively. In this review, we will summarize the current status of IGF-1R molecular imaging in cancer, in which 5 major classes of ligands that have been developed for noninvasive IGF-1R molecular imaging will be discussed: natural ligands, monoclonal antibodies, antibody fragments, affibodies, and small molecules. For decades, IGF-1R molecular imaging is studied in full swing and more effort is needed in the future.
Collapse
Affiliation(s)
- Yingying Sun
- 1 Molecular Imaging Research Center, Harbin Medical University, Harbin, Heilongjiang, China.,2 TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xilin Sun
- 1 Molecular Imaging Research Center, Harbin Medical University, Harbin, Heilongjiang, China.,2 TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,3 Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Baozhong Shen
- 1 Molecular Imaging Research Center, Harbin Medical University, Harbin, Heilongjiang, China.,2 TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
16
|
Rodriguez-Monterrosas C, Diaz-Aragon R, Cortes-Reynosa P, Salazar EP. Linoleic acid induces an increased response to insulin in MDA-MB-231 breast cancer cells. J Cell Biochem 2018; 119:5413-5425. [PMID: 29363790 DOI: 10.1002/jcb.26694] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/22/2018] [Indexed: 12/12/2022]
Abstract
Epidemiological studies and animal models suggest a link between high levels of dietary fat intake and an increased risk of developing breast cancer. Hyperinsulinemia is a feature of obesity, diabetes, and metabolic syndrome that is associated with an increased breast cancer risk. Insulin is a hormone involved in metabolic regulation of carbohydrate. However, it is also a growth factor that mediates proliferation and migration. Linoleic acid (LA) is a fatty acid that induces migration and invasion in breast cancer cells. In the present study, we demonstrate, for the first time, that treatment with LA increases IR and IGF1R expression through a Free Fatty Acid Receptor 4 (FFAR4)-, lipooxygenases (LOXs)-, and SRC-dependent pathway in MDA-MB-231 breast cancer cells, and similarly induces an increase of IR expression in MCF-7 breast cancer cells. In addition, insulin induces tyrosine phosphorylation of IR/IGF1R and migration in MDA-MB-231 cells pretreated with LA, whereas it augments the increase in migration in MCF-7 cells pretreated with LA. Pretreatment of MDA-MB-231 cells with LA induces invasion, proliferation, and increase the MMP-9 secretion induced by insulin. In summary, our findings demonstrate that treatment with LA induces a higher response to insulin in breast cancer cells.
Collapse
Affiliation(s)
| | | | | | - Eduardo P Salazar
- Departamento de Biologia Celular, Cinvestav-IPN, Mexico City, Mexico
| |
Collapse
|
17
|
Vishwamitra D, George SK, Shi P, Kaseb AO, Amin HM. Type I insulin-like growth factor receptor signaling in hematological malignancies. Oncotarget 2018; 8:1814-1844. [PMID: 27661006 PMCID: PMC5352101 DOI: 10.18632/oncotarget.12123] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 09/12/2016] [Indexed: 12/19/2022] Open
Abstract
The insulin-like growth factor (IGF) signaling system plays key roles in the establishment and progression of different types of cancer. In agreement with this idea, substantial evidence has shown that the type I IGF receptor (IGF-IR) and its primary ligand IGF-I are important for maintaining the survival of malignant cells of hematopoietic origin. In this review, we discuss current understanding of the role of IGF-IR signaling in cancer with a focus on the hematological neoplasms. We also address the emergence of IGF-IR as a potential therapeutic target for the treatment of different types of cancer including plasma cell myeloma, leukemia, and lymphoma.
Collapse
Affiliation(s)
- Deeksha Vishwamitra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Suraj Konnath George
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ping Shi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Ahmed O Kaseb
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hesham M Amin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
18
|
Schvartsman G, Gutierrez-Barrera AM, Song J, Ueno NT, Peterson SK, Arun B. Association between weight gain during adjuvant chemotherapy for early-stage breast cancer and survival outcomes. Cancer Med 2017; 6:2515-2522. [PMID: 29024537 PMCID: PMC5673950 DOI: 10.1002/cam4.1207] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/10/2017] [Accepted: 08/25/2017] [Indexed: 12/13/2022] Open
Abstract
Obese and overweight women have an increased risk of breast cancer and worse outcomes at the time of diagnosis. Women tend to gain weight after breast cancer diagnosis and during chemotherapy for early‐stage disease, which may in turn increase risk for worse outcomes. We examined if weight gained during adjuvant chemotherapy was associated with worse survival outcomes. We queried our database for data on patients who received adjuvant third‐generation chemotherapy for early‐stage breast cancer. Univariate and multivariate analyses by Cox regression were performed for survival outcomes across three categories according to BMI variation from start to end of chemotherapy: >0.5 kg/m2 loss or gain and stable BMI (±0.5 kg/m2). We included 1998 patients in this study. Women over 50 years old and postmenopausal were more likely to lose weight during adjuvant chemotherapy, whereas women under 30 years old gained more weight (P < 0.001). At 1 year postchemotherapy, patients tended to return to their original weight (ρ = −0.3, P < 0.001). On multivariate analysis, BMI increase of >0.5 kg/m2 compared to maintaining BMI was marginally associated with increased locoregional recurrence risk (HR: 2.53; 95% CI, 1.18–5.45; P = 0.017), adjusting for grade, stage, and radiation delivery. Weight variation during adjuvant chemotherapy for early‐stage breast cancer may occur as both weight gain and weight loss in a balanced manner. Furthermore, this variation seems to be transient in nature and does not appear to significantly influence recurrence rates and overall survival.
Collapse
Affiliation(s)
- Gustavo Schvartsman
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Juhee Song
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Susan K Peterson
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas.,MD Anderson Cancer Center, Houston, Texas
| | - Banu Arun
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
19
|
Shlomai G, Zelenko Z, Antoniou IM, Stasinopoulos M, Tobin-Hess A, Vitek MP, LeRoith D, Gallagher EJ. OP449 inhibits breast cancer growth without adverse metabolic effects. Endocr Relat Cancer 2017; 24:519-529. [PMID: 28830934 PMCID: PMC5678946 DOI: 10.1530/erc-17-0077] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 07/21/2017] [Indexed: 12/23/2022]
Abstract
Hyperinsulinemia is associated with a decrease in breast cancer recurrence-free survival and overall survival. Inhibition of insulin receptor signaling is associated with glycemic dysregulation. SET is a direct modulator of PP2A, which negatively regulates the PI3K/AKT/mTOR pathway. OP449, a SET inhibitor, decreases AKT/mTOR activation. The effects of OP449 treatment on breast cancer growth in the setting of pre-diabetes, and its metabolic implications are currently unknown. We found that the volumes and weights of human MDA-MB-231 breast cancer xenografts were greater in hyperinsulinemic mice compared with controls (P < 0.05), and IR phosphorylation was 4.5-fold higher in these mice (P < 0.05). Human and murine breast cancer tumors treated with OP449 were 47% and 39% smaller than controls (P < 0.05, for both, respectively). AKT and S6RP phosphorylation were 82% and 34% lower in OP449-treated tumors compared with controls (P < 0.05, P = 0.06, respectively). AKT and S6RP phosphorylation in response to insulin was 30% and 12% lower in cells, pre-treated with OP449, compared with control cells (P < 0.01, P < 0.05, respectively). However, even with decreased AKT/mTOR activation, body weights and composition, blood glucose and plasma insulin, glucose tolerance, serum triglyceride and cholesterol levels were similar between OP449-treated mice and controls. Xenografts and liver tissue from OP449-treated mice showed a 64% and 70% reduction in STAT5 activation, compared with controls (P < 0.01 and P = 0.06, respectively). Our data support an anti-neoplastic effect of OP449 on human breast cancer cells in vitro and in xenografts in the setting of hyperinsulinemia. OP449 led to the inhibition of AKT/mTOR signaling, albeit, not leading to metabolic derangements.
Collapse
Affiliation(s)
- Gadi Shlomai
- Division of EndocrinologyDiabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- The Dr Pinchas Borenstein Talpiot Medical Leadership Program 2013Tel-Hashomer, Israel
| | - Zara Zelenko
- Division of EndocrinologyDiabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Irini Markella Antoniou
- Division of EndocrinologyDiabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Marilyn Stasinopoulos
- Division of EndocrinologyDiabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Aviva Tobin-Hess
- Division of EndocrinologyDiabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Michael P Vitek
- CognosciInc., Durham, North Carolina, USA
- Department of NeurologyDuke University Medical Center, Research Drive, Durham, North Carolina, USA
| | - Derek LeRoith
- Division of EndocrinologyDiabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Emily Jane Gallagher
- Division of EndocrinologyDiabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
20
|
The Mechanisms and Effects of Physical Activity on Breast Cancer. Clin Breast Cancer 2017; 17:272-278. [PMID: 28233686 DOI: 10.1016/j.clbc.2017.01.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/13/2017] [Indexed: 12/15/2022]
Abstract
Breast cancer is the most common cancer in women. An extensive part of this health problem can be prevented by an active lifestyle. Physical activity can reduce the risk of breast cancer, reduce the rate of recurrence, and increase the survival rate of patients with breast cancer. The aim of this review was to summarize our current knowledge regarding the effects of physical activity on breast cancer risk, recurrence, and survival. Furthermore, we investigated 5 possible underlying mechanisms through which physical activity has an influence on breast cancer (ie, a reduction of sex hormones, metabolic hormones, adipokines and oxidative stress, and an improvement of the immune function). In this review, we give a complete overview of this subject.
Collapse
|
21
|
Cohen-Sinai T, Cohen Z, Werner H, Berger R. Identification of BRCA1 As a Potential Biomarker for Insulin-Like Growth Factor-1 Receptor Targeted Therapy in Breast Cancer. Front Endocrinol (Lausanne) 2017; 8:148. [PMID: 28706506 PMCID: PMC5489552 DOI: 10.3389/fendo.2017.00148] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 06/12/2017] [Indexed: 01/05/2023] Open
Abstract
The insulin-like growth factor-1 receptor (IGF1R) emerged in recent years as a promising therapeutic target in oncology. Identification of potential biomarkers capable of predicting response to IGF1R-targeted therapy is of cardinal importance. Tumor suppressor BRCA1 has important roles in multiple pathways, including gene transcription, DNA damage repair, and control of apoptosis. Early studies have identified the IGF1R gene as a downstream target for inhibitory regulation by wild-type, but not mutant, BRCA1. The aim of the present study was to evaluate the hypothesis that the mutational status of BRCA1 may influence the ability of IGF1R-directed therapies to efficiently inhibit the IGF1R axis. Using breast cancer-derived cell lines expressing a wild-type or a mutant BRCA1, we demonstrate that the capacity of MK-0646, a monoclonal antibody antagonist to the human IGF1R, to inhibit insulin-like growth factor-1-stimulated IGF1R and downstream mediators' phosphorylation was impaired in mutant BRCA1-expressing cell lines. In addition, the antibody was able to reduce proliferation of wild-type BRCA1-expressing cells but had a reduced inhibitory effect in mutant BRCA1-expressing cells. In summary, our data indicate that the mutational status of BRCA1 must be taken into account when selecting patients for IGF1R targeting protocols.
Collapse
Affiliation(s)
- Tali Cohen-Sinai
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Zoya Cohen
- Institute of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Yoran Institute for Human Genome Research, Tel Aviv University, Tel Aviv, Israel
- *Correspondence: Haim Werner,
| | - Raanan Berger
- Institute of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
22
|
Park YS, Kim DJ, Koo H, Jang SH, You YM, Cho JH, Yang SJ, Yu ES, Jung Y, Lee DC, Kim JA, Park ZY, Park KC, Yeom YI. AKT-induced PKM2 phosphorylation signals for IGF-1-stimulated cancer cell growth. Oncotarget 2016; 7:48155-48167. [PMID: 27340866 PMCID: PMC5217008 DOI: 10.18632/oncotarget.10179] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 06/04/2016] [Indexed: 12/14/2022] Open
Abstract
Pyruvate kinase muscle type 2 (PKM2) exhibits post-translational modifications in response to various signals from the tumor microenvironment. Insulin-like growth factor 1 (IGF-1) is a crucial signal in the tumor microenvironment that promotes cell growth and survival in many human cancers. Herein, we report that AKT directly interacts with PKM2 and phosphorylates it at Ser-202, which is essential for the nuclear translocation of PKM2 protein under stimulation of IGF-1. In the nucleus, PKM2 binds to STAT5A and induces IGF-1-stimulated cyclin D1 expression, suggesting that PKM2 acts as an important factor inducing STAT5A activation under IGF-1 signaling. Concordantly, overexpression of STAT5A in cells deficient in PKM2 expression failed to restore IGF-induced growth, whereas reconstitution of PKM2 in PKM2 knockdown cells restored the IGF-induced growth capacity. Our findings suggest a novel role of PKM2 in promoting the growth of cancers with dysregulated IGF/phosphoinositide 3-kinase/AKT signaling.
Collapse
Affiliation(s)
- Young Soo Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon 34113, Korea
| | - Dong Joon Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Han Koo
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon 34113, Korea
| | - Se Hwan Jang
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
| | - Yeon-Mi You
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon 34113, Korea
| | - Jung Hee Cho
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Suk-Jin Yang
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Eun Sil Yu
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Yuri Jung
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Dong Chul Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Jung-Ae Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon 34113, Korea
| | - Zee-Yong Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
| | - Kyung Chan Park
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Young Il Yeom
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon 34113, Korea
| |
Collapse
|
23
|
Choi SI, Kim SY, Lee J, Cho EW, Kim IG. TM4SF4 overexpression in radiation-resistant lung carcinoma cells activates IGF1R via elevation of IGF1. Oncotarget 2015; 5:9823-37. [PMID: 25344917 PMCID: PMC4259440 DOI: 10.18632/oncotarget.2450] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Transmembrane 4 L six family member 4 (TM4SF4) is a member of the tetraspanin L6 domain family. Other members of this family, TM4SF1 (also known as L6-Ag) and TM4SF5, have been shown to be upregulated in multiple tumors and involved in epithelial-to-mesenchymal transition and cell migration. However, unlike its homologs, little is known about TM4SF4. Here, we show that TM4SF4 was highly expressed in radiation-resistant lung adenocarcinoma cells, such as A549 and Calu-3 cells, and its expression activated cell growth, migration, and invasion. Overexpression of TM4SF4 in A549 cells increased the activation of PI3K, AKT, and NF-kappaB and the expression of PTEN. IGF1R was clearly activated by overexpression of TM4SF4, although EGFR was also slightly activated. TM4SF4 expression was correlated with the increased expression of IGF1, consequently resulting in IGF1R activation. Tumorigenic activity of TM4SF4 in lung adenocarcinoma cells was also demonstrated by xenograft assay; however, this activity was almost completely suppressed by treatment with anti-TM4SF4 antibody. Our results suggest that TM4SF4 overexpression in lung carcinoma cells results in resistance to radiotherapy via IGF1-induced IGF1R activation and blocking the activity of TM4SF4 using specific antibody can be a promising therapeutics against TM4SF4-overexpressing lung adenocarcinoma.
Collapse
Affiliation(s)
- Soo-Im Choi
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute (KAERI), Daedeok-daero, Yuseong-gu, Daejeon, South Korea. Department of Radiation Biotechnology and Applied Radioisotope, Korea University of Science and Technology, Daedeok-daero, Yuseong-gu, Daejeon, South Korea
| | - Seo-Yeon Kim
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute (KAERI), Daedeok-daero, Yuseong-gu, Daejeon, South Korea
| | - Jaeha Lee
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute (KAERI), Daedeok-daero, Yuseong-gu, Daejeon, South Korea. Department of Radiation Biotechnology and Applied Radioisotope, Korea University of Science and Technology, Daedeok-daero, Yuseong-gu, Daejeon, South Korea
| | - Eun-Wie Cho
- Epigenomics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Gwahak-ro, Yuseong-gu, Daejeon, South Korea
| | - In-Gyu Kim
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute (KAERI), Daedeok-daero, Yuseong-gu, Daejeon, South Korea. Department of Radiation Biotechnology and Applied Radioisotope, Korea University of Science and Technology, Daedeok-daero, Yuseong-gu, Daejeon, South Korea
| |
Collapse
|
24
|
Quandt Z, Flom JD, Tehranifar P, Reynolds D, Terry MB, McDonald JA. The association of alcohol consumption with mammographic density in a multiethnic urban population. BMC Cancer 2015; 15:1094. [PMID: 25777420 PMCID: PMC4374505 DOI: 10.1186/s12885-015-1094-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 02/20/2015] [Indexed: 12/21/2022] Open
Abstract
Background Alcohol consumption is associated with higher breast cancer risk. While studies suggest a modest association between alcohol intake and mammographic density, few studies have examined the association in racial/ethnic minority populations. Methods We assessed dense breast area and total breast area from digitized film mammograms in an urban cohort of African American (42%), African Caribbean (22%), white (22%), and Hispanic Caribbean (9%) women (n = 189, ages 40-61). We examined the association between alcohol intake and mammographic density (percent density and dense area). We used linear regression to examine mean differences in mammographic density across alcohol intake categories. We considered confounding by age, body mass index (BMI), hormone contraceptive use, family history of breast cancer, menopausal status, smoking status, nativity, race/ethnicity, age at first birth, and parity. Results Fifty percent currently consumed alcohol. Women who consumed >7 servings/week of alcohol, but not those consuming ≤7 servings/week, had higher percent density compared to nondrinkers after full adjustments (servings/week >7 β = 8.2, 95% Confidence Interval (CI) 1.8, 14.6; ≤7 β = -0.5, 95% CI -3.7, 2.8). There was a positive association between high alcohol intake and dense area after full adjustments (servings/week >7 β = 5.8, 95% CI -2.7, 14.2; ≤7 β = -0.1, 95% CI -4.4, 4.2). We did not observe race/ethnicity modification of the association between alcohol intake and percent density. In women with a BMI of <25 kg/m2, drinkers consuming >7 servings/week of alcohol had a 17% increase in percent density compared to nondrinkers (95% CI 5.4, 29.0) and there was no association in women with a BMI ≥ 25 kg/m2 (BMI ≥ 25-30 kg/m2 > 7 β = 5.1, 95% CI -8.5, 18.7 and BMI > 30 kg/m2 > 7 β = 0.5, 95% CI -6.5, 7.5) after adjusting for age and BMI (continuous). Conclusion In a racially/ethnically diverse cohort, women who consumed >7 servings/week of alcohol, especially those with a BMI < 25 kg/m2, had higher percent density.
Collapse
Affiliation(s)
- Zoe Quandt
- Department of Epidemiology, Columbia University Medical Center, Mailman School of Public Health, New York, NY, USA,
| | | | | | | | | | | |
Collapse
|
25
|
Zhang X, Zhang Y, Yu Y, Liu J, Yuan Y, Zhao Y, Li H, Wang J, Wang Z. Convergence and divergence of genetic and modular networks between diabetes and breast cancer. J Cell Mol Med 2015; 19:1094-102. [PMID: 25752479 PMCID: PMC4420611 DOI: 10.1111/jcmm.12504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/22/2014] [Indexed: 12/03/2022] Open
Abstract
Diabetes mellitus (DM) and breast cancer (BC) can simultaneously occur in the same patient populations, but the molecular relationship between them remains unknown. In this study, we constructed genetic networks and used modularized analysis approaches to investigate the multi-dimensional characteristics of two diseases and one disease subtype. A text search engine (Agilent Literature Search 2.71) and MCODE software were applied to validate potential subnetworks and to divide the modules, respectively. A total of 793 DM-related genes, 386 type 2 diabetes (T2DM) genes and 873 BC-related genes were identified from the Online Mendelian Inheritance in Man database. For DM and BC, a total of 99 overlapping genes, 9 modules, 29 biological processes and 7 pathways were identified. Meanwhile, for T2DM and BC, 56 overlapping genes, 5 modules, 20 biological processes and 12 pathways were identified. Based on the Gene Ontology functional enrichment analysis of the top 10 non-overlapping modules of the two diseases, 10 biological functions and 5 pathways overlapped between them. The glycosphingolipid and lysosome pathways verified molecular mechanisms of cell death related to both DM and BC. We also identified new biological functions of dopamine receptors and four signalling pathways (Parkinson's disease, Alzheimer's disease, Huntington's disease and long-term depression) related to both diseases; these warrant further investigation. Our results illustrate the landscape of the novel molecular substructures between DM and BC, which may support a new model for complex disease classification and rational therapies for multiple diseases.
Collapse
Affiliation(s)
- Xiaoxu Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Rota LM, Wood TL. Crosstalk of the Insulin-Like Growth Factor Receptor with the Wnt Signaling Pathway in Breast Cancer. Front Endocrinol (Lausanne) 2015; 6:92. [PMID: 26106366 PMCID: PMC4460810 DOI: 10.3389/fendo.2015.00092] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 05/17/2015] [Indexed: 12/21/2022] Open
Abstract
The insulin-like growth factor system has long been considered a pathway that promotes cell proliferation, survival, and transformation, and is thus a promoter of tumorigenesis. However, recent failure of clinical trials for IGF-1R inhibitors reveals the need for a better understanding of how this pathway functions in specific tumor subtypes. Ongoing studies are designed to uncover biomarkers and downstream targets to enhance therapeutic strategies. Other approaches in specific tumor models reveal complex interactions between IGF signaling and other tumor initiating pathways. Here, we review relevant background and recent studies suggesting that inhibiting the IGF-1R can amplify Wnt and Notch signaling pathways in a model of triple negative breast cancer.
Collapse
Affiliation(s)
- Lauren M. Rota
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School and Cancer Center, Rutgers University, Newark, NJ, USA
| | - Teresa L. Wood
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School and Cancer Center, Rutgers University, Newark, NJ, USA
- *Correspondence: Teresa L. Wood, Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School and Cancer Center, Rutgers University, 205 South Orange Avenue, Newark, NJ 07101, USA,
| |
Collapse
|
27
|
Abstract
AbstractBreast cancer is one of the most common types of cancer among women worldwide. Epidemiological and experimental studies confirm the beneficial effects of physical exercise both in patients who have suffered a relapse and in those who are in the course of a treatment. Researchers suggest that regular physical effort performed 3-5 times a week at 20-40 MET reduces the risk of development of breast cancer. Scientific evidence suggests that aerobic training can be safe and effective even during the oncological treatment, which, depending on the stage of the disease, may consist of surgical treatment combined with chemotherapy, radiotherapy or hormone therapy. In addition, it significantly improves the function of the circulatory and respiratory systems, reduces fatigue and improves the quality of life of patients struggling with the disease. There are many theories on the mechanisms that are triggered by aerobic workout. Despite the numerous studies on this subject, which have been conducted over the years, we still cannot say how physical activity reduces the risk of development of breast cancer. The most frequently mentioned biological mechanisms include the influence of oestrogen, metabolic hormones, growth factor, inflammatory markers, response from the immune system, insulin resistance and oxidative stress. Many studies into the process of carcinogenesis are carried out in laboratory conditions on animals. Due to application of different experimental models, the results of such studies are often ambiguous. Nevertheless, scientists see a huge potential in this kind of experiments, because some of them have already attested to the protective effects of physical exercise against the appearance of breast cancer, but also they have increased our understanding of the processes through which this risk is reduced. Perhaps, in the future physical activity will become a part of a cancer treatment thanks to the results of such studies.
Collapse
|
28
|
Pabalan NA, Seim I, Jarjanazi H, Chopin LK. Associations between ghrelin and ghrelin receptor polymorphisms and cancer in Caucasian populations: a meta-analysis. BMC Genet 2014; 15:118. [PMID: 25376984 PMCID: PMC4228186 DOI: 10.1186/s12863-014-0118-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 10/22/2014] [Indexed: 12/15/2022] Open
Abstract
Background There is growing evidence that the ghrelin axis, including ghrelin (GHRL) and its receptor, the growth hormone secretagogue receptor (GHSR), play a role in cancer progression. Ghrelin gene and ghrelin receptor gene polymorphisms have been reported to have a range of effects in cancer, from increased risk, to protection from cancer, or having no association. In this study we aimed to clarify the role of ghrelin and ghrelin receptor polymorphisms in cancer by performing a meta-analysis of published case–control studies. We conducted searches of the literature published up to January 2013 in MEDLINE using the PubMed search engine. Individual data on 8,430 cases and 14,008 controls from six case–control studies of an all Caucasian population were evaluated for three ghrelin gene (GHRL; rs696217, rs4684677, rs2075356) and one ghrelin receptor (GHSR; rs572169) polymorphism in breast cancer, esophageal cancer, colorectal cancer and non-Hodgkins lymphoma. Results In the overall analysis, homozygous and recessive associations indicated that the minor alleles of rs696217 and rs2075356 GHRL polymorphisms conferred reduced cancer risk (odds ratio [OR] 0.61-0.78). The risk was unchanged for breast cancer patients when analysed separately (OR 0.73-0.83). In contrast, the rs4684677 GHRL and the rs572169 GHSR polymorphisms conferred increased breast cancer risk (OR 1.97-1.98, p = 0.08 and OR 1.42-1.43, p = 0.08, respectively). All dominant and co-dominant effects showed null effects (OR 0.96-1.05), except for the rs572169 co-dominant effect, with borderline increased risk (OR 1.08, p = 0.05). Conclusions This study suggests that the rs696217 and rs2075356 ghrelin gene (GHRL) polymorphisms may protect carriers against breast cancer, and the rs4684677 GHRL and rs572169 GHSR polymorphisms may increase the risk among carriers. In addition, larger studies are required to confirm these findings.
Collapse
|
29
|
Liu X, Giguère V. Inactivation of RARβ inhibits Wnt1-induced mammary tumorigenesis by suppressing epithelial-mesenchymal transitions. NUCLEAR RECEPTOR SIGNALING 2014; 12:e004. [PMID: 25422594 PMCID: PMC4242291 DOI: 10.1621/nrs.12004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/18/2014] [Indexed: 12/20/2022]
Abstract
Retinoic acid receptor β (RARβ) has been proposed to act as a tumor suppressor in
breast cancer. In contrast, recent data have shown that RARβ promotes ERBB2-induced mammary
gland tumorigenesis through remodeling of the stromal compartment and activation of
cancer-associated fibroblasts. However, it is currently unknown whether RARβ oncogenic
activity is specific to ERBB2-induced tumors, or whether it influences the initiation and
progression of other breast cancer subtypes. Accordingly, we set out to investigate the involvement
of RARβ in basal-like breast cancer using mouse mammary tumor virus (MMTV)-wingless-related
integration site 1 (Wnt1)-induced mammary gland tumorigenesis as a model system. We found that
compared with wild type mice, inactivation of Rarb resulted in a lengthy delay in
Wnt1-induced mammary gland tumorigenesis and in a significantly slower tumor growth
rate. Ablation of Rarb altered the composition of the stroma, repressed the
activation of cancer-associated fibroblasts, and reduced the recruitment of inflammatory cells and
angiogenesis. Reduced expression of IGF-1 and activity of its downstream signaling pathway
contribute to attenuate EMT in the Rarb-null tumors. Our results show that, in the
absence of retinoid signaling via RARβ, reduced IGF-1 signaling results in suppression of
epithelial-mesenchymal transition and delays tumorigenesis induced by the Wnt1
oncogene. Accordingly, our work reinforces the concept that antagonizing RARβ-dependent
retinoid signaling could provide a therapeutic avenue to treat poor outcome breast cancers.
Collapse
Affiliation(s)
- Xingxing Liu
- Goodman Cancer Research Centre, 1160 Pine Avenue West, McGill University, Montréal, Québec H3A 1A3 (XL, VG) and Departments of Biochemistry, Medicine and Oncology, 3655 Promenade Sir William Osler, McGill University, Montréal, Québec H3G 1Y6 (VG), Canada
| | - Vincent Giguère
- Goodman Cancer Research Centre, 1160 Pine Avenue West, McGill University, Montréal, Québec H3A 1A3 (XL, VG) and Departments of Biochemistry, Medicine and Oncology, 3655 Promenade Sir William Osler, McGill University, Montréal, Québec H3G 1Y6 (VG), Canada
| |
Collapse
|
30
|
Wiseman AJ, Lynch BM, Cameron AJ, Dunstan DW. Associations of change in television viewing time with biomarkers of postmenopausal breast cancer risk: the Australian Diabetes, Obesity and Lifestyle Study. Cancer Causes Control 2014; 25:1309-19. [DOI: 10.1007/s10552-014-0433-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 07/03/2014] [Indexed: 01/24/2023]
|
31
|
Yen YC, Lin C, Lin SW, Lin YS, Weng SF. Effect of metformin on the incidence of head and neck cancer in diabetics. Head Neck 2014; 37:1268-73. [PMID: 24801563 DOI: 10.1002/hed.23743] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 03/26/2014] [Accepted: 05/03/2014] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The purpose of this study was to examine the effect of metformin on head and neck cancer in patients with diabetes. METHODS We compared 66,600 patients, all with diabetes and all newly diagnosed with head and neck cancer in 2002. Half were being treated with metformin for diabetes (Met(+) ) and half were not (Met(-) : controls). All were matched for comorbidities (obesity, coronary artery disease, hyperlipidemia, and hypertension), sex, and age. The risk of head and neck cancer at the end of 2011 was determined. RESULTS The incidence of head and neck cancer was 34% lower in the Met(+) cohort than in the Met(-) cohort (adjusted hazard ratio [HR] = 0.66; 95% confidence interval [CI] = 0.55-0.79). The risks for oropharyngeal cancer (adjusted HR = 0.66; 95% CI = 0.17-0.74) and nasopharyngeal carcinoma (NPC; adjusted HR = 0.50; 95% CI = 0.31-0.80) were significantly lower in the Met(+) cohort than in the Met(-) cohort. CONCLUSION Metformin is associated with a lower risk of developing head and neck cancer in patients with diabetes.
Collapse
Affiliation(s)
- Yung-Chang Yen
- Department of Ophthalmology, Chi Mei Medical Centre, Liou-Ying, Tainan City, Taiwan
| | - Charlene Lin
- Department of Molecular and Cell Biology, University of California, Berkeley, California
| | - Shih-Wei Lin
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Yung-Song Lin
- Department of Otolaryngology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Otolaryngology, Chi Mei Medical Center, Tainan, Taiwan
| | - Shih-Feng Weng
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan.,Department of Hospital and Health Care Administration, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
| |
Collapse
|
32
|
Franco L, Williams FMK, Trofimov S, Malkin I, Surdulescu G, Spector T, Livshits G. Assessment of age-related changes in heritability and IGF-1 gene effect on circulating IGF-1 levels. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9622. [PMID: 24493200 PMCID: PMC4082604 DOI: 10.1007/s11357-014-9622-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 01/20/2014] [Indexed: 05/19/2023]
Abstract
It is well established that insulin-like growth factor 1 (IGF-1) circulating levels correlate with age and that heritability and influence of IGF-1 gene variation on IGF-1 levels also well-known. However, the influence of age on the genetic factors determining IGF-1 levels is not clear. In this study, we compared heritability estimates between younger (<52 years) and older (>52 years) twins and tested: (a) whether single nucleotide polymorphisms (SNPs) lying within 100 kbp of the IGF-1 gene are also associated with IGF-1 variation and (b) whether associated SNPs show interaction with age on IGF-1 levels. To achieve these aims, we measured plasma levels of IGF-1 and genotyped 18 SNPs with minor allele frequency >0.1 in a large sample, 4,471 UK female twins. Heritability explained 42 % of IGF-1 variation adjusted for age and in unadjusted sample was independent of age. Ten SNPs in four haploblocks showed significant association with IGF-1 levels, with p = 0.01-0.0005. The most distal SNP was located up to 90 kbp from the IGF-1 gene. When their age-dependent effects were examined, one SNP, rs855203, showed significant (p = 0.0009) age-dependent interaction effect on IGF-1 levels variation. This is the first study to test the age × genotype interaction in IGF-1 levels. The genomic region marked by rs855203 may consequently be of significance for further molecular and pharmacogenetic research, in particular in advanced age.
Collapse
Affiliation(s)
- Liran Franco
- />Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Frances M. K. Williams
- />Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK
| | - Svetlana Trofimov
- />Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ida Malkin
- />Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gabriela Surdulescu
- />Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK
| | - Timothy Spector
- />Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK
| | - Gregory Livshits
- />Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- />Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK
- />Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978 Israel
| |
Collapse
|
33
|
Fine EJ, Segal-Isaacson C, Herzkopf S, Sparano J, Romano M, Feinman R, Tomuta N, Bontempo A, Negassa A. Can inhibiting insulin/IGF signaling with dietary carbohydrate restriction play a role in treatment/prevention of cancers? Cancer Metab 2014. [PMCID: PMC4073060 DOI: 10.1186/2049-3002-2-s1-o31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
34
|
Song MJ, Lee EJ, Yang Y, Sung MK. Luteolin supplementation modulates mammary tumor growth in C3H mice fed diet with high- and low-fat content. Nutr Cancer 2013; 66:523-30. [PMID: 24074002 DOI: 10.1080/01635581.2013.780629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
In this study we investigated the effects of luteolin supplementation (0.05% w/w) on mammary tumor growth in C3H mice, a strain of mouse mammary tumor virus negative, fed either high-fat (45% fat of energy) or low-fat diet (15% fat of energy). Animals (n = 12/group) were allocated into 4 experimental groups (low-fat diet, low-fat diet + luteolin supplementation, high-fat diet, high-fat diet + luteolin supplementation). Experimental diet were fed for 13 wk and 7,12-dimethylbenz[a]anthracene was administered once a week for 6 wk starting at Week 1 to induce mammary tumors. Study results showed that animals on low-fat diet supplemented with luteolin exhibited longer tumor latency and lower tumor weights and sizes compared to the other groups. Animals fed high-fat diet showed increased serum IGF-1 levels and the elevated mammary tissue expression of Ki-67, IRS-1, pp38, Cdk4, and Cdk6. Luteolin inhibited IRS-1, Cdk4, and Cdk6 expression in high-fat fed animals. The expression of pp38, cyclinD1, and Bcl-xL was suppressed by luteolin supplementation both in the low-fat and high-fat diet groups. These results suggest that excess energy supply increases the risk of mammary tumor formation and luteolin suppresses tumor formation regardless of dietary fat content through its cell cycle regulatory and proapoptotic activity.
Collapse
Affiliation(s)
- Min-Ji Song
- a Department of Food and Nutrition , Sookmyung Women's University , Seoul , Korea
| | | | | | | |
Collapse
|
35
|
Risk of cancer in diabetes: the effect of metformin. ISRN ENDOCRINOLOGY 2013; 2013:636927. [PMID: 24224094 PMCID: PMC3800579 DOI: 10.1155/2013/636927] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 08/16/2013] [Indexed: 12/20/2022]
Abstract
Cancer is the second cause of death. Association of diabetes as a growing and costly disease with cancer is a major health concern. Meanwhile, preexisting diabetes is associated with an increased risk of all-cause and cancer-specific mortalities. Presence of diabetes related comorbidities, poorer response to cancer treatment, and excess mortality related to diabetes are among the most important explanations. Although diabetes appear to be a risk factor for cancer and is associated with the mortality risk in cancer patients, several factors such as diabetes duration, multiple drug therapy, and the presence of diabetes comorbidities make the assessment of the effect of diabetes treatment on cancer risk and mortality difficult. Metformin is the drug of choice for the treatment of type 2 diabetes. The available evidence from basic science, clinical, and population-based research supports the anticancer effect of metformin. However, randomized controlled clinical trials do not provide enough evidence for a strong protective effect of metformin on cancer incidence or mortality. One of the most important limitations of these trials is the short duration of the followup. Further long-term randomized controlled clinical trials specifically designed to determine metformin effect on cancer risk are needed to provide the best answer to this challenge.
Collapse
|
36
|
Ezzat S, Zheng L, Florez JC, Stefan N, Mayr T, Hliang MM, Jablonski K, Harden M, Stančáková A, Laakso M, Haring HU, Ullrich A, Asa SL. The cancer-associated FGFR4-G388R polymorphism enhances pancreatic insulin secretion and modifies the risk of diabetes. Cell Metab 2013; 17:929-940. [PMID: 23747250 PMCID: PMC4005358 DOI: 10.1016/j.cmet.2013.05.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 01/18/2013] [Accepted: 04/05/2013] [Indexed: 12/11/2022]
Abstract
The fibroblast growth factor receptor 4 (FGFR4)-R388 single-nucleotide polymorphism has been associated with cancer risk and prognosis. Here we show that the FGFR4-R388 allele yields a receptor variant that preferentially promotes STAT3/5 signaling. This STAT activation transcriptionally induces Grb14 in pancreatic endocrine cells to promote insulin secretion. Knockin mice with the FGFR4 variant allele develop pancreatic islets that secrete more insulin, a feature that is reversed through Grb14 deletion and enhanced with FGF19 administration. We also show in humans that the FGFR4-R388 allele enhances islet function and may protect against type 2 diabetes. These data support a common genetic link underlying cancer and hyperinsulinemia.
Collapse
Affiliation(s)
- Shereen Ezzat
- Ontario Cancer Institute, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - Lei Zheng
- Ontario Cancer Institute, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Jose C Florez
- Center for Human Genetic Research and Diabetes Research Center (Diabetes Unit), Massachusetts General Hospital, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02142, USA
| | | | - Thomas Mayr
- Department of Molecular Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Maw Maw Hliang
- Ontario Cancer Institute, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Kathleen Jablonski
- The Biostatistics Center, George Washington University, Rockville, MD 20852, USA
| | - Maegan Harden
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alena Stančáková
- Department of Medicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland, 70210 Kuopio, Finland
| | | | - Axel Ullrich
- Department of Molecular Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Sylvia L Asa
- Ontario Cancer Institute, University Health Network, Toronto, ON M5G 2M9, Canada
| |
Collapse
|
37
|
Seim I, Lubik AA, Lehman ML, Tomlinson N, Whiteside EJ, Herington AC, Nelson CC, Chopin LK. Cloning of a novel insulin-regulated ghrelin transcript in prostate cancer. J Mol Endocrinol 2013; 50:179-91. [PMID: 23267039 DOI: 10.1530/jme-12-0150] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ghrelin is a multifunctional hormone, with roles in stimulating appetite and regulating energy balance, insulin secretion and glucose homoeostasis. The ghrelin gene locus (GHRL) is highly complex and gives rise to a range of novel transcripts derived from alternative first exons and internally spliced exons. The wild-type transcript encodes a 117 amino acid preprohormone that is processed to yield the 28 amino acid peptide ghrelin. Here, we identified insulin-responsive transcription corresponding to cryptic exons in intron 2 of the human ghrelin gene. A transcript, termed in2c-ghrelin (intron 2-cryptic), was cloned from the testis and the LNCaP prostate cancer cell line. This transcript may encode an 83 amino acid preproghrelin isoform that codes for ghrelin, but not obestatin. It is expressed in a limited number of normal tissues and in tumours of the prostate, testis, breast and ovary. Finally, we confirmed that in2c-ghrelin transcript expression, as well as the recently described in1-ghrelin transcript, is significantly upregulated by insulin in cultured prostate cancer cells. Metabolic syndrome and hyperinsulinaemia have been associated with prostate cancer risk and progression. This may be particularly significant after androgen deprivation therapy for prostate cancer, which induces hyperinsulinaemia, and this could contribute to castrate-resistant prostate cancer growth. We have previously demonstrated that ghrelin stimulates prostate cancer cell line proliferation in vitro. This study is the first description of insulin regulation of a ghrelin transcript in cancer and should provide further impetus for studies into the expression, regulation and function of ghrelin gene products.
Collapse
Affiliation(s)
- Inge Seim
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Kelvin Grove, Brisbane, Queensland 4059, Australia
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The insulin-like growth factor (IGF) system has a direct effect on cellular proliferation and survival, and interacts with genetic and environmental factors implicated in causing cancer. Experimental, clinical, and epidemiological evidence show that the IGF signalling pathways are important mediators in the biochemical and molecular chain of events that lead from a phenotypically normal cell to one harbouring neoplastic traits. BRCA1 and BRCA2 have an important role in the development of hereditary and sporadic breast and ovarian cancer. Recent evidence suggests that risk of cancer conferred by BRCA mutations can be modified by genetic and environmental factors, including ambient concentrations of IGF-1 and polymorphisms in IGF system components. This Review addresses interactions between the IGF and BRCA1 signalling pathways, and emphasises the convergence of IGF-1-mediated cell survival, proliferative pathways, and BRCA1-mediated tumour protective pathways. Understanding the complex interactions between these signalling pathways might improve our understanding of basic molecular oncology processes and help to identify new molecular targets, predictive biomarkers, and approaches for optimising cancer therapies.
Collapse
Affiliation(s)
- Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | | |
Collapse
|
39
|
Rock CL, Pande C, Flatt SW, Ying C, Pakiz B, Parker BA, Williams K, Bardwell WA, Heath DD, Nichols JF. Favorable changes in serum estrogens and other biologic factors after weight loss in breast cancer survivors who are overweight or obese. Clin Breast Cancer 2013; 13:188-95. [PMID: 23375717 DOI: 10.1016/j.clbc.2012.12.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 11/21/2012] [Accepted: 12/08/2012] [Indexed: 01/09/2023]
Abstract
BACKGROUND Obesity is associated with an increased risk for recurrence and all-cause mortality in breast cancer survivors. Excess adiposity is associated with increased estrogen, insulin, and leptin, and with decreased sex hormone binding globulin (SHBG) concentrations, which may promote breast cancer progression and recurrence. This study aimed to assess the effects of weight loss on these factors. PATIENTS AND METHODS Breast cancer survivors who were overweight or obese (n = 220) and who were enrolled in a weight loss intervention study provided baseline and follow-up blood samples and weight data. Serum estrogens, SHBG, insulin, and leptin were measured at baseline, 6 months, and 18 months. RESULTS Weight loss of ≥5% of initial weight decreased leptin and insulin compared with those who did not achieve that amount of weight loss (P < .0001). Weight loss also increased SHBG at 6 and 18 months (P < .01). Postmenopausal women who lost ≥5% of body weight at 6 months had lower estrone (P = .02), estradiol (P = .002), and bioavailable estradiol (P = .001) concentrations than women who did not lose at least 5% of body weight, and weight loss at 18 months was significantly related to a change in serum bioavailable estradiol concentration (P = .02). CONCLUSIONS Favorable changes in estrogens, SHBG, insulin, and leptin were observed in association with weight loss in these women who were overweight or obese and who had been diagnosed and treated for breast cancer. Weight loss appears to have favorable effects on hormonal and biologic factors associated with increased risk for recurrence and poorer prognosis.
Collapse
Affiliation(s)
- Cheryl L Rock
- Department of Family and Preventive Medicine and Cancer Prevention and Control Program, University of California, San Diego, La Jolla, CA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Rock CL, Byers TE, Colditz GA, Demark-Wahnefried W, Ganz PA, Wolin KY, Elias A, Krontiras H, Liu J, Naughton M, Pakiz B, Parker BA, Sedjo RL, Wyatt H. Reducing breast cancer recurrence with weight loss, a vanguard trial: the Exercise and Nutrition to Enhance Recovery and Good Health for You (ENERGY) Trial. Contemp Clin Trials 2012; 34:282-95. [PMID: 23266440 DOI: 10.1016/j.cct.2012.12.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 12/13/2012] [Accepted: 12/17/2012] [Indexed: 01/21/2023]
Abstract
Breast cancer is the most common invasive cancer among women in developed countries. Obesity is a major risk factor for breast cancer recurrence and mortality in both pre- and postmenopausal women. Co-morbid medical conditions are common among breast cancer survivors. The Exercise and Nutrition to Enhance Recovery and Good Health for You (ENERGY) study is a 4-year randomized clinical trial of 693 overweight/obese women aged ≥21years diagnosed with any early stage breast cancer (stages I[≥1cm]-III) within the previous five years, designed to demonstrate the feasibility of achieving sustained weight loss and to examine the impact of weight loss on quality of life and co-morbidities, and to enable future exploration of biochemical mechanisms linking obesity to lower likelihood of disease-free survival. This trial is strategically designed as a vanguard for a fully-powered trial of women who will be evaluated for breast cancer recurrence and disease-free survival. Participants were recruited between 2010 and 2012 at four sites, had completed initial therapies, and had a body mass index between 25 and 45kg/m(2). The intervention featured a group-based cognitive-behavioral weight loss program with telephone counseling and tailored newsletters to support initial weight loss and subsequent maintenance, with the goal of 7% weight loss at two years. This study has high potential to have a major impact on clinical management and outcomes after a breast cancer diagnosis. This trial initiates the effort to establish weight loss support for overweight or obese breast cancer survivors as a new standard of clinical care.
Collapse
|
41
|
Neurotrophins and their receptors in breast cancer. Cytokine Growth Factor Rev 2012; 23:357-65. [DOI: 10.1016/j.cytogfr.2012.06.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 06/06/2012] [Indexed: 12/21/2022]
|
42
|
Patterson RE, Rock CL, Kerr J, Natarajan L, Marshall SJ, Pakiz B, Cadmus-Bertram LA. Metabolism and breast cancer risk: frontiers in research and practice. J Acad Nutr Diet 2012; 113:288-96. [PMID: 23127511 DOI: 10.1016/j.jand.2012.08.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 08/06/2012] [Indexed: 10/27/2022]
Abstract
Fifty years ago the causes of cancer were largely unknown. Since then, it has become clear that a strong relationship exists between obesity and many cancers, particularly postmenopausal breast cancer. A major challenge in understanding the link between obesity and cancer risk has been elucidating the biological basis underlying the association. Although this remains unresolved, the main candidate systems linking adiposity and cancer risk are insulin and the insulin-like growth factor-1 axis, endogenous reproductive hormones, and chronic inflammation. Our purpose is to provide a mechanistic overview of the hypothesized relationship between diet, physical activity, and obesity with breast cancer risk and progression. In addition, we will provide examples of recently funded randomized clinical trials examining metabolic risk factors in relation to breast cancer risk and survival. Additional research is warranted to validate the strength and consistency of the relationships among diet, these biomarkers, and breast cancer risk. As these relationships become clearer, future studies will be needed to develop effective intervention programs to prevent breast cancer and improve cancer prognosis by promoting a healthy lifestyle.
Collapse
Affiliation(s)
- Ruth E Patterson
- Department of Family and Preventive Medicine, Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA 92093-0901, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Baxi SM, Tan W, Murphy ST, Smeal T, Yin MJ. Targeting 3-phosphoinoside-dependent kinase-1 to inhibit insulin-like growth factor-I induced AKT and p70 S6 kinase activation in breast cancer cells. PLoS One 2012; 7:e48402. [PMID: 23119004 PMCID: PMC3485233 DOI: 10.1371/journal.pone.0048402] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 09/25/2012] [Indexed: 12/15/2022] Open
Abstract
Binding of IGF to IGF-IR activates PI3K to generate PIP3 which in turn recruits and activates proteins that contain a pleckstrin homology (PH) domain, including AKT and PDK1. PDK1 is highly expressed in breast tumor samples and breast cancer cell lines. Here we demonstrate that targeting PDK1 with the potent and selective PDK1 inhibitor PF-5177624 in the IGF-PI3K pathway blocks breast cancer cell proliferation and transformation. Breast cancer cell lines MCF7 and T47D, representing the luminal ER positive subtype and harboring PIK3CA mutations, were most responsive to IGF-I induction resulting in upregulated AKT and p70S6K phosphorylation via PDK1 activation. PF-5177624 downregulated AKT and p70S6K phosphorylation, blocked cell cycle progression, and decreased cell proliferation and transformation to block IGFR-I induced activation in breast cancer cells. These results may provide insight into clinical strategies for developing an IGFR-I inhibitor and/or a PDK1 inhibitor in luminal breast cancer patients.
Collapse
Affiliation(s)
- Sangita M. Baxi
- Oncology Research, Pfizer Worldwide Research and Development, San Diego, California, United States of America
| | - Wei Tan
- Oncology Research, Pfizer Worldwide Research and Development, San Diego, California, United States of America
| | - Sean T. Murphy
- Medicinal Chemistry, Pfizer Worldwide Research and Development, San Diego, California, United States of America
| | - Tod Smeal
- Oncology Research, Pfizer Worldwide Research and Development, San Diego, California, United States of America
| | - Min-Jean Yin
- Oncology Research, Pfizer Worldwide Research and Development, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
44
|
Chen Y, Zhu C, Peng Z, Dai Y, Gu Y. Lentivirus-mediated short-hairpin RNA targeting IGF-1R inhibits growth and lymphangiogenesis in breast cancer. Oncol Rep 2012; 28:1778-84. [PMID: 22895778 DOI: 10.3892/or.2012.1964] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 07/17/2012] [Indexed: 11/05/2022] Open
Abstract
In this study, we investigated the effects of lentivirus (LV)-mediated short hairpin RNA (shRNA) targeting IGF-1R on the growth and lymphangiogenesis of breast cancer. The LV vector effectively delivered the IGF-1R shRNA to MDA-MB‑231 cells, leading to significant reduction of IGF-1R mRNA and protein expression. Infection of MDA-MB-231 cells with LV-IGF-1R shRNA reduced cell growth and migration. Transplantation of MDA-MB-231 cells with suppressed IGF-1R expression in SCID mice reduced tumor growth and lymphangiogenesis. These data collectively suggest that LV-mediated shRNA is an effective way to suppress IGF-1R expression and to inhibit growth and lymphangiogenesis of breast cancer. Specific inhibition of IGF-1R expression with shRNA represents a promising approach for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yaning Chen
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, HuangPu District, Shanghai 200011, PR China
| | | | | | | | | |
Collapse
|
45
|
Fine EJ, Segal-Isaacson CJ, Feinman RD, Herszkopf S, Romano MC, Tomuta N, Bontempo AF, Negassa A, Sparano JA. Targeting insulin inhibition as a metabolic therapy in advanced cancer: a pilot safety and feasibility dietary trial in 10 patients. Nutrition 2012; 28:1028-35. [PMID: 22840388 DOI: 10.1016/j.nut.2012.05.001] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 04/06/2012] [Accepted: 05/11/2012] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Most aggressive cancers demonstrate a positive positron emission tomographic (PET) result using ¹⁸F-2-fluoro-2-deoxyglucose (FDG), reflecting a glycolytic phenotype. Inhibiting insulin secretion provides a method, consistent with published mechanisms, for limiting cancer growth. METHODS Eligible patients with advanced incurable cancers had a positive PET result, an Eastern Cooperative Oncology Group performance status of 0 to 2, normal organ function without diabetes or recent weight loss, and a body mass index of at least 20 kg/m². Insulin inhibition, effected by a supervised carbohydrate dietary restriction (5% of total kilocalories), was monitored for macronutrient intake, body weight, serum electrolytes, β-hydroxybutyrate, insulin, and insulin-like growth factors-1 and -2. An FDG-PET scan was obtained at study entry and exit. RESULTS Ten subjects completed 26 to 28 d of the study diet without associated unsafe adverse effects. Mean caloric intake decreased 35 ± 6% versus baseline, and weight decreased by a median of 4% (range 0.0-6.1%). In nine patients with prior rapid disease progression, five with stable disease or partial remission on PET scan after the diet exhibited a three-fold higher dietary ketosis than those with continued progressive disease (n = 4, P = 0.018). Caloric intake (P = 0.65) and weight loss (P = 0.45) did not differ in those with stable disease or partial remission versus progressive disease. Ketosis was associated inversely with serum insulin levels (P = 0.03). CONCLUSION Preliminary data demonstrate that an insulin-inhibiting diet is safe and feasible in selected patients with advanced cancer. The extent of ketosis, but not calorie deficit or weight loss, correlated with stable disease or partial remission. Further study is needed to assess insulin inhibition as complementary to standard cytotoxic and endocrine therapies.
Collapse
Affiliation(s)
- Eugene J Fine
- Department of Radiology (Nuclear Medicine), Albert Einstein College of Medicine, Bronx, New York, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Roop RP, Ma CX. Endocrine resistance in breast cancer: molecular pathways and rational development of targeted therapies. Future Oncol 2012; 8:273-92. [PMID: 22409464 DOI: 10.2217/fon.12.8] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Endocrine resistance presents a major challenge in the management of estrogen receptor (ER)-positive breast cancer and is an area under intense investigation. Although the underlying mechanism is still poorly understood, many studies point towards the 'cross-talk' between ER and growth factor receptor signaling pathways as the key in the development of estrogen-independent growth in breast cancer. This review aims to provide the reader our current understanding of various molecular pathways that mediate endocrine resistance and that are being evaluated as therapeutic targets for ER-positive breast cancer. While most of the agents that target these pathways have only been tested in Phase I or small Phase II trials, some have shown encouraging results. A critical issue that remains is the development of research strategies and clinical trials that take into account the molecular heterogeneity of ER-positive breast cancer.
Collapse
Affiliation(s)
- Ryan P Roop
- Washington University School of Medicine in Saint Louis, Department of Medicine, Divisions of Hematology & Oncology, St Louis, MO, USA
| | | |
Collapse
|
47
|
A pilot safety-feasibility dietary trial targeting insulin inhibition in ten patients with advanced cancer. BMC Proc 2012. [PMCID: PMC3374260 DOI: 10.1186/1753-6561-6-s3-p60] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
48
|
Insulin-like growth factor-I regulates GPER expression and function in cancer cells. Oncogene 2012; 32:678-88. [PMID: 22430216 DOI: 10.1038/onc.2012.97] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Functional cross talk between insulin-like growth factor-I (IGF-I) system and estrogen signaling has been largely reported, although the underlying molecular mechanisms remain to be fully elucidated. As GPR30/GPER mediates rapid cell responses to estrogens, we evaluated the potential of IGF-I to regulate GPER expression and function in estrogen receptor (ER)α-positive breast (MCF-7) and endometrial (Ishikawa) cancer cells. We found that IGF-I transactivates the GPER promoter sequence and upregulates GPER mRNA and protein levels in both cells types. Similar data were found, at least in part, in carcinoma-associated fibroblasts. The upregulation of GPER expression by IGF-I involved the IGF-IR/PKCδ/ERK/c-fos/AP1 transduction pathway and required ERα, as ascertained by specific pharmacological inhibitors and gene-silencing. In both MCF-7 and Ishikawa cancer cells, the IGF-I-dependent cell migration required GPER and its main target gene CTGF, whereas the IGF-I-induced proliferation required both GPER and cyclin D1. Our data demonstrate that the IGF-I system regulates GPER expression and function, triggering the activation of a signaling network that leads to the migration and proliferation of cancer cells.
Collapse
|
49
|
Fidler MJ, Shersher DD, Borgia JA, Bonomi P. Targeting the insulin-like growth factor receptor pathway in lung cancer: problems and pitfalls. Ther Adv Med Oncol 2012; 4:51-60. [PMID: 22423264 PMCID: PMC3296080 DOI: 10.1177/1758834011427576] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The insulin-like growth factor (IGF) pathway is a complex pathway involving interactions between membrane-bound receptors, ligands, binding proteins, downstream effectors, and other receptor tyrosine kinase signaling cascades. The IGF pathway has been identified as a potential therapeutic target in non-small cell lung cancer (NSCLC) based on the following provocative factors. Preclinical observations in NSCLC have shown that this pathway is involved in tumor cell proliferation, survival, and invasiveness. In addition, IGF-1R protein expression is found in a significant number of non-small cell tumor specimens. Initial therapeutic efforts involved the development of monoclonal antibodies and tyrosine kinase inhibitors that target IGF-1R, a transmembrane receptor tyrosine kinase. Enthusiasm for targeting this pathway increased when a randomized phase II study showed that combining an anti-IGF-1R monoclonal antibody (figitumumab) with a platinum doublet resulted in a higher response rate and trends for superior progression-free survival and overall survival. Subsequently, a phase III study failed to confirm the promising results observed in the phase II trial. Currently, investigators are studying different monoclonal antibodies and tyrosine kinases targeting IGF-1R. In unselected patients, results presented thus far do not suggest efficacy of this agent. However, retrospective subgroup analyses suggest that circulating IGF-1 levels might identify patients who could benefit from treatment with an IGF-1R monoclonal antibody and may warrant further exploratory studies for predictive molecular markers. The purpose of this paper is to briefly discuss the IGF pathway and its relationship with other signaling pathways in lung cancer and to review the ongoing IGF clinical trials and efforts to identify predictive molecular markers.
Collapse
Affiliation(s)
- Mary Jo Fidler
- Section of Medical Oncology, Rush University Medical Center, 1725 West Harrison Street, Suite 821, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
50
|
Liu BY, Soloviev I, Huang X, Chang P, Ernst JA, Polakis P, Sakanaka C. Mammary tumor regression elicited by Wnt signaling inhibitor requires IGFBP5. Cancer Res 2012; 72:1568-78. [PMID: 22307840 DOI: 10.1158/0008-5472.can-11-3668] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Wnt ligand-driven tumor growth is inhibited by the soluble Wnt inhibitor Fzd8CRD, but the mechanism through which this effect is mediated is unknown. In the MMTV-Wnt1 mouse model, regression of mammary tumors by Fzd8CRD treatment coincides with an acute and strong induction of insulin-like growth factor (IGF)-binding protein IGFBP5, an antagonist of IGF signaling that mediates involution of mammary gland in females after offspring are weaned. In this study, we show that repression of this IGF inhibitory pathway is crucial for Wnt-driven growth of mammary tumors. We found that IGFBP5 regulation was mediated by the β-catenin-dependent Wnt pathway. Wnt, in addition to IGF ligands, facilitated tumor growth by paracrine communication among tumor cells. In addition, Fzd8CRD caused precocious induction of IGFBP5 in normal mammary glands undergoing involution, implying an acceleration of the involution process by inhibition of Wnt signaling. The molecular and phenotypic parallel between tumor regression and mammary gland involution suggests that Wnt-driven mammary tumors use the same growth mechanism as proliferating normal mammary glands.
Collapse
Affiliation(s)
- Bob Y Liu
- Department of Cancer Targets, Genentech Inc., South San Francisco, California 94080, USA
| | | | | | | | | | | | | |
Collapse
|