1
|
Matias MA, Sharma N. Nonsurgical Management of High-Risk Lesions. Radiol Clin North Am 2024; 62:679-686. [PMID: 38777542 DOI: 10.1016/j.rcl.2023.12.005] [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] [Indexed: 05/25/2024]
Abstract
This article highlights the recent publications and changing trends in practice regarding management of high-risk lesions of the breast. Traditional management has always been a surgical operation but this is recognized as overtreatment. It is recognized that overdiagnosis is inevitable but what we can control is overtreatment. Vacuum-assisted excision is now established as an alternative technique to surgery for further sampling of these high-risk lesions in the United Kingdom. Guidelines from the United Kingdom and Europe now recognize this alternative pathway, and data are available showing that vacuum-assisted excision is a safe alternative to surgery.
Collapse
Affiliation(s)
- Mariana Afonso Matias
- Breast Unit, Leeds Teaching Hospital NHS Trust, Level 1 Chancellor Wing, St James Hospital, Beckett Street, Leeds LS9 7TF
| | - Nisha Sharma
- Breast Unit, Leeds Teaching Hospital NHS Trust, Level 1 Chancellor Wing, St James Hospital, Beckett Street, Leeds LS9 7TF.
| |
Collapse
|
2
|
Rubio IT, Wyld L, Marotti L, Athanasiou A, Regitnig P, Catanuto G, Schoones JW, Zambon M, Camps J, Santini D, Dietz J, Sardanelli F, Varga Z, Smidt M, Sharma N, Shaaban AM, Gilbert F. European guidelines for the diagnosis, treatment and follow-up of breast lesions with uncertain malignant potential (B3 lesions) developed jointly by EUSOMA, EUSOBI, ESP (BWG) and ESSO. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:107292. [PMID: 38061151 DOI: 10.1016/j.ejso.2023.107292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 01/16/2024]
Abstract
INTRODUCTION Breast lesions of uncertain malignant potential (B3) include atypical ductal and lobular hyperplasias, lobular carcinoma in situ, flat epithelial atypia, papillary lesions, radial scars and fibroepithelial lesions as well as other rare miscellaneous lesions. They are challenging to categorise histologically, requiring specialist training and multidisciplinary input. They may coexist with in situ or invasive breast cancer (BC) and increase the risk of subsequent BC development. Management should focus on adequate classification and management whilst avoiding overtreatment. The aim of these guidelines is to provide updated information regarding the diagnosis and management of B3 lesions, according to updated literature review evidence. METHODS These guidelines provide practical recommendations which can be applied in clinical practice which include recommendation grade and level of evidence. All sections were written according to an updated literature review and discussed at a consensus meeting. Critical appraisal by the expert writing committee adhered to the 23 items in the international Appraisal of Guidelines, Research and Evaluation (AGREE) tool. RESULTS Recommendations for further management after core-needle biopsy (CNB) or vacuum-assisted biopsy (VAB) diagnosis of a B3 lesion reported in this guideline, vary depending on the presence of atypia, size of lesion, sampling size, and patient preferences. After CNB or VAB, the option of vacuum-assisted excision or surgical excision should be evaluated by a multidisciplinary team and shared decision-making with the patient is crucial for personalizing further treatment. De-escalation of surgical intervention for B3 breast lesions is ongoing, and the inclusion of vacuum-assisted excision (VAE) will decrease the need for surgical intervention in further approaches. Communication with patients may be different according to histological diagnosis, presence or absence of atypia, or risk of upgrade due to discordant imaging. Written information resources to help patients understand these issues alongside with verbal communication is recommended. Lifestyle interventions have a significant impact on BC incidence so lifestyle interventions need to be suggested to women at increased BC risk as a result of a diagnosis of a B3 lesion. CONCLUSIONS These guidelines provide a state-of-the-art overview of the diagnosis, management and prognosis of B3 lesions in modern multidisciplinary breast practice.
Collapse
Affiliation(s)
- Isabel T Rubio
- Breast Surgical Oncology, Clinica Universidad de Navarra, Madrid, Spain; European Society of Breast Cancer Specialists (EUSOMA), Florence, Italy; European Society of Surgical Oncology (ESSO), Brussels, Belgium.
| | - Lynda Wyld
- Department of Oncology and Metabolism, University of Sheffield Medical School, Beech Hill Road, Sheffield, S10 2RX, UK; Doncaster and Bassetlaw Teaching Hospitals NHS Foundation Trust, Doncaster, UK
| | - Lorenza Marotti
- European Society of Breast Cancer Specialists (EUSOMA), Florence, Italy
| | | | - Peter Regitnig
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Giuseppe Catanuto
- Humanitas-Istituto Clinico Catanese Misterbianco, Italy; Fondazione G.Re.T.A., ETS, Napoli, Italy
| | - Jan W Schoones
- Research Policy & Graduate School Advisor, Leiden University Medical Center Leiden, the Netherlands
| | - Marzia Zambon
- Europa Donna - The European Breast Cancer Coalition, Milan, Italy
| | - Julia Camps
- Breast Health Units in Ribera Salud Hospitals.Valencia, Spain
| | - Donatella Santini
- Department of Pathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - Jill Dietz
- The American Society of Breast Surgeons, Columbia, MD, USA
| | - Francesco Sardanelli
- Department of Biomedical Sciences for Health, Università Degli Studi di Milano, Milan, Italy; Unit of Radiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Zsuzsanna Varga
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Marjolein Smidt
- GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands; Department of Surgery, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Nisha Sharma
- Breast Unit, Level 1 Chancellor Wing, St James Hospital, Beckett Street Leeds, West Yorkshire, LS9 7TF, UK
| | - Abeer M Shaaban
- Cellular Pathology, Queen Elizabeth Hospital Birmingham, Birmingham, UK; Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Fiona Gilbert
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, UK.
| |
Collapse
|
3
|
Dewidar SA, Hamdy O, Soliman MM, El Gayar AM, El-Mesery M. Enhanced therapeutic efficacy of doxorubicin/cyclophosphamide in combination with pitavastatin or simvastatin against breast cancer cells. Med Oncol 2023; 41:7. [PMID: 38051378 PMCID: PMC10697881 DOI: 10.1007/s12032-023-02248-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/11/2023] [Indexed: 12/07/2023]
Abstract
Fighting breast tumors mandates finding different agents devoid of chemotherapy side effects. Repurposing existing drugs, such as statins, presents a promising avenue for the development of novel cancer therapeutics. Based on the different effects of statin members, this study aims to evaluate the effect of two of the most promising lipophilic statins, Simvastatin and Pitavastatin, and their combination with a conventional chemotherapeutic regimen of doxorubicin and cyclophosphamide on breast cancer cells. MDA-MB-231 and MCF7 cell lines were used to analyze the effects of Pitavastatin and simvastatin in combination with doxorubicin/cyclophosphamide. Cell viability and cell cycle were analyzed and certain apoptosis-related genes such as Bax, Bcl2, and caspase-3, besides cyclin D1 were analyzed using qPCR. The viability of breast cancer cells decreased significantly after treatment with a doxorubicin/cyclophosphamide combination in the presence of Pitavastatin or simvastatin compared with dual doxorubicin/cyclophosphamide with a higher effect in MDA-MB-231 cells than MCF7. In MDA-MB-231, The triple combination of Pitavastatin or simvastatin with doxorubicin/cyclophosphamide resulted in an increase in the expression levels of apoptotic markers than treatment with doxorubicin/cyclophosphamide combination (Bax (p-value = 0.09& 0.02, respectively), Bax/Bcl2 ratio (p-value = 0.0002& <0.0001, respectively)). However, the increase in caspase3 wasn't significant (p-value = 0.45& 0.09, respectively). Moreover, the expression of cyclin D1 decreased (p-value = 0.0002& <0.0001, respectively) and the cell cycle was arrested in the G1 phase. Combination of Pitavastatin or simvastatin with doxorubicin/ cyclophosphamide may induce apoptosis in breast cancer cells via upregulation of the Bax/Bcl2 pathway, potentially providing a promising new therapeutic strategy for breast cancer.
Collapse
Affiliation(s)
- Samar A Dewidar
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Omar Hamdy
- Surgical oncology department, Oncology Center, Mansoura University, Mansoura, Egypt.
| | - Moetaza M Soliman
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Amal M El Gayar
- Biochemistry Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohamed El-Mesery
- Biochemistry Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
4
|
Abstract
To identify regulators of triple-negative breast cancer (TNBC), gene expression profiles of malignant parts of TNBC (mTNBC) and normal adjacent (nadj) parts of the same breasts have been compared. We are interested in the roles of estrogen receptor β (ERβ) and the cytochrome P450 family (CYPs) as drivers of TNBC. We examined by RNA sequencing the mTNBC and nadj parts of five women. We found more than a fivefold elevation in mTNBC of genes already known to be expressed in TNBC: BIRC5/survivin, Wnt-10A and -7B, matrix metalloproteinases (MMPs), chemokines, anterior gradient proteins, and lysophosphatidic acid receptor and the known basal characteristics of TNBC, sox10, ROPN1B, and Col9a3. There were two unexpected findings: 1) a strong induction of CYPs involved in activation of fatty acids (CYP4), and in inactivation of calcitriol (CYP24A1) and retinoic acid (CYP26A1); and 2) a marked down-regulation of FOS, FRA1, and JUN, known tethering partners of ERβ. ERβ is expressed in 20 to 30% of TNBCs and is being evaluated as a target for treating TNBC. We used ERβ+ TNBC patient-derived xenografts in mice and found that the ERβ agonist LY500703 had no effect on growth or proliferation. Expression of CYPs was confirmed by immunohistochemistry in formalin-fixed and paraffin-embedded (FFPE) TNBC. In TNBC cell lines, the CYP4Z1-catalyzed fatty acid metabolite 20-hydroxyeicosatetraenoic acid (20-HETE) increased proliferation, while calcitriol decreased proliferation but only after inhibition of CYP24A1. We conclude that CYP-mediated pathways can be drivers of TNBC but that ERβ is unlikely to be a tumor suppressor because the absence of its main tethering partners renders ERβ functionless on genes involved in proliferation and inflammation.
Collapse
|
5
|
Kryczyk-Poprawa A, Zupkó I, Bérdi P, Żmudzki P, Piotrowska J, Pękala E, Berdys A, Muszyńska B, Opoka W. Photodegradation of Bexarotene and Its Implication for Cytotoxicity. Pharmaceutics 2021; 13:pharmaceutics13081220. [PMID: 34452181 PMCID: PMC8401567 DOI: 10.3390/pharmaceutics13081220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 11/24/2022] Open
Abstract
A detailed understanding of the stability of an active pharmaceutical ingredient and a pharmaceutical dosage form is essential for the drug-development process and for safe and effective use of medicines. Photostability testing as an inherent part of stability studies provides valuable knowledge on degradation pathways and structures of products generated under UV irradiation. Photostability is particularly important for topically administered drugs, as they are more exposed to UV radiation. Bexarotene is a more recent third-generation retinoid approved by the U.S. Food and Drug Administration and the European Medicines Agency as a topically applied anticancer agent. The present study aimed to assess bexarotene photostability, including the presence of UV filters, which have been permitted to be used in cosmetic products in Europe and the USA. The bexarotene photostability testing was performed in ethanol solutions and in formulations applied on PMMA plates. The UPLC-MS/MS technique was used to determine the tested substance. The presence of photocatalysts such as TiO2 or ZnO, as well as the organic UV filters avobenzone, benzophenone-3, meradimate, and homosalate, could contribute to degradation of bexarotene under UV irradiation. Four photocatalytic degradation products of bexarotene were identified for the first time. The antiproliferative properties of the degradation products of bexarotene were assessed by MTT assay on a panel of human adherent cancer cells, and concentration-dependent growth inhibition was evidenced on all tested cell lines. The cytotoxicity of the formed products after 4 h of UV irradiation was significantly higher than that of the parent compound (p < 0.05). Furthermore non-cancerous murine fibroblasts exhibited marked concentration-dependent inhibition by bexarotene, while the degradation products elicited more pronounced antiproliferative action only at the highest applied concentration.
Collapse
Affiliation(s)
- Agata Kryczyk-Poprawa
- Department of Inorganic and Analytical Chemistry, Jagiellonian University Medical College, 30-688 Kraków, Poland; (J.P.); (W.O.)
- Correspondence:
| | - István Zupkó
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, H-6720 Szeged, Hungary; (I.Z.); (P.B.)
- Interdisciplinary Centre for Natural Products, University of Szeged, H-6720 Szeged, Hungary
| | - Péter Bérdi
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, H-6720 Szeged, Hungary; (I.Z.); (P.B.)
| | - Paweł Żmudzki
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 30-688 Kraków, Poland;
| | - Joanna Piotrowska
- Department of Inorganic and Analytical Chemistry, Jagiellonian University Medical College, 30-688 Kraków, Poland; (J.P.); (W.O.)
| | - Elżbieta Pękala
- Department of Pharmaceutical Biochemistry, Jagiellonian University Medical College, 30-688 Kraków, Poland;
| | | | - Bożena Muszyńska
- Department of Pharmaceutical Botany, Jagiellonian University Collegium Medicum, 30-688 Kraków, Poland;
| | - Włodzimierz Opoka
- Department of Inorganic and Analytical Chemistry, Jagiellonian University Medical College, 30-688 Kraków, Poland; (J.P.); (W.O.)
| |
Collapse
|
6
|
Baraya YS, Yankuzo HM, Wong KK, Yaacob NS. Strobilanthes crispus bioactive subfraction inhibits tumor progression and improves hematological and morphological parameters in mouse mammary carcinoma model. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113522. [PMID: 33127562 DOI: 10.1016/j.jep.2020.113522] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Locally known as 'pecah batu', 'bayam karang', 'keci beling' or 'batu jin', the Malaysian medicinal herb, Strobilanthes crispus (S. crispus), is traditionally used by the local communities as alternative or adjuvant remedy for cancer and other ailments and to boost the immune system. S. crispus has demonstrated multiple anticancer therapeutic potential in vitro and in vivo. A pharmacologically active fraction of S. crispus has been identified and termed as F3. Major constituents profiled in F3 include lutein and β-sitosterol. AIM OF THE STUDY In this study, the effects of F3, lutein and β-sitosterol on tumor development and metastasis were investigated in 4T1-induced mouse mammary carcinoma model. MATERIALS AND METHODS Tumor-bearing mice were fed with F3 (100 mg/kg/day), lutein (50 mg/kg/day) and β-sitosterol (50 mg/kg/day) for 30 days (n = 5 each group). Tumor physical growth parameters, animal body weight and development of secondary tumors were investigated. The safety profile of F3 was assessed using hematological and histomorphological changes on the major organs in normal control mice (NM). RESULTS Our findings revealed significant reduction of physical tumor growth parameters in all tumor-bearing mice treated with F3 (TM-F3), lutein (TM-L) or β-sitosterol (TM-β) as compared with the untreated group (TM). Statistically significant reduction in body weight was observed in TM compared to the NM or treated (TM-F3, TM-L and TM-β) groups. Histomorphological examination of tissue sections from the F3-treated group showed normal features of the vital organs (i.e., liver, kidneys, lungs and spleen) which were similar to those of NM. Administration of F3 to NM mice (NM-F3) did not cause significant changes in full blood count values. CONCLUSION F3 significantly reduced the total tumor burden and prevented secondary tumor development in metastatic breast cancer without significant toxicities in 4T1-induced mouse mammary carcinoma model. The current study provides further support for therapeutic development of F3 with further pharmacokinetics studies.
Collapse
Affiliation(s)
- Yusha'u Shu'aibu Baraya
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Usmanu Danfodiyo University, Sokoto, Nigeria.
| | - Hassan Muhammad Yankuzo
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria.
| | - Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kelantan, Malaysia.
| | - Nik Soriani Yaacob
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kelantan, Malaysia.
| |
Collapse
|
7
|
Bytautaite M, Petrikaite V. Comparative Study of Lipophilic Statin Activity in 2D and 3D in vitro Models of Human Breast Cancer Cell Lines MDA-MB-231 and MCF-7. Onco Targets Ther 2020; 13:13201-13209. [PMID: 33380809 PMCID: PMC7769197 DOI: 10.2147/ott.s283033] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Introduction Statins are a type of drugs that are used to lower cholesterol level in blood. Since the early 1990s, it has been known that statins could be beneficial in cancer therapy. However, data remain controversial, especially regarding estrogen receptors status. Despite many studies in breast cancer models in vitro, the correlations of effects of separate statins in various model systems remain unclear. Aim Our aim was to evaluate the anticancer activity of lovastatin, mevastatin, pitavastatin and simvastatin on different subtypes of human breast cancer (MDA-MB-231 and MCF-7 cell lines) in spatially different 2D and 3D cultures in vitro. Materials and Methods Cell viability was tested using MTT assay. Effect of statins on cell colony formation was evaluated by calculating breast cancer cell colony area and number. The effect on cell migration was estimated by “wound healing” assay. The activity of compounds in 3D cultures was evaluated by measuring the spheroid size changes during incubation. Results Among the tested statins, pitavastatin had the greatest effect on the viability of breast cancer MDA-MB-231 and MCF-7 cell lines. The mevastatin and pitavastatin mostly decreased the MDA-MB-231 and MCF-7 cell colony formation. All statins at 90% of their estimated effective concentration (EC50) and simvastatin at 10% of its EC50 concentration suppressed the MCF-7 cells migration in “wound healing” assay. Only higher concentrations of mevastatin and pitavastatin slowed down the MDA-MB-231 cell migration. Statins showed different activity on 3D cell cultures growth. Lovastatin and simvastatin delayed the growth of MDA-MB-231 cell spheroids, while mevastatin and pitavastatin reduced the growth of MCF-7 spheroids. Conclusion Statins possess different anticancer activity in human breast cancer MDA-MB-231 and MCF-7 cell cultures. Pitavastatin and simvastatin showed the highest activity in most tested assays, especially against MCF-7 cell line.
Collapse
Affiliation(s)
- Meda Bytautaite
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, LT-50162, Lithuania
| | - Vilma Petrikaite
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, LT-50162, Lithuania
| |
Collapse
|
8
|
Yang C, Yu H, Chen R, Tao K, Jian L, Peng M, Li X, Liu M, Liu S. CXCL1 stimulates migration and invasion in ER‑negative breast cancer cells via activation of the ERK/MMP2/9 signaling axis. Int J Oncol 2019; 55:684-696. [PMID: 31322183 PMCID: PMC6685590 DOI: 10.3892/ijo.2019.4840] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 07/01/2019] [Indexed: 12/19/2022] Open
Abstract
Chemokine (C‑X‑C motif) ligand 1 (CXCL1), a member of the CXC chemokine family, has been reported to be a critical factor in inflammatory diseases and tumor progression; however, its functions and molecular mechanisms in estrogen receptor α (ER)‑negative breast cancer (BC) remain largely unknown. The present study demonstrated that CXCL1 was upregulated in ER‑negative BC tissues and cell lines compared with ER‑positive tissues and cell lines. Treatment with recombinant human CXCL1 protein promoted ER‑negative BC cell migration and invasion in a dose‑dependent manner, and stimulated the activation of phosphorylated (p)‑ extracellular signal‑regulated kinase (ERK)1/2, but not p‑STAT3 or p‑AKT. Conversely, knockdown of CXCL1 in BC cells attenuated these effects. Additionally, CXCL1 increased the expression of matrix metalloproteinase (MMP)2/9 via the ERK1/2 pathway. Inhibition of MEK1/2 by its antagonist U0126 reversed the effects of CXCL1 on MMP2/9 expression. Furthermore, immunohistochemical analysis revealed a strong positive association between CXCL1 and p‑ERK1/2 expression levels in BC tissues. In conclusion, the present study demonstrated that CXCL1 is highly expressed in ER‑negative BC, and stimulates BC cell migration and invasion via the ERK/MMP2/9 pathway. Therefore, CXCL1 may serve as a potential therapeutic target in ER‑negative BC.
Collapse
Affiliation(s)
- Chengcheng Yang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Haochen Yu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Rui Chen
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Kai Tao
- Department of the Second of Gynecology Oncology, Shanxi Provincial Tumor Hospital, The Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shanxi 710061, P.R. China
| | - Lei Jian
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Meixi Peng
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiaotian Li
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Manran Liu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Shengchun Liu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
9
|
Focus on Cdc42 in Breast Cancer: New Insights, Target Therapy Development and Non-Coding RNAs. Cells 2019; 8:cells8020146. [PMID: 30754684 PMCID: PMC6406589 DOI: 10.3390/cells8020146] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/30/2019] [Accepted: 02/08/2019] [Indexed: 12/25/2022] Open
Abstract
Breast cancer is the most common malignant tumors in females. Although the conventional treatment has demonstrated a certain effect, some limitations still exist. The Rho guanosine triphosphatase (GTPase) Cdc42 (Cell division control protein 42 homolog) is often upregulated by some cell surface receptors and oncogenes in breast cancer. Cdc42 switches from inactive guanosine diphosphate (GDP)-bound to active GTP-bound though guanine-nucleotide-exchange factors (GEFs), results in activation of signaling cascades that regulate various cellular processes such as cytoskeletal changes, proliferation and polarity establishment. Targeting Cdc42 also provides a strategy for precise breast cancer therapy. In addition, Cdc42 is a potential target for several types of non-coding RNAs including microRNAs and lncRNAs. These non-coding RNAs is extensively involved in Cdc42-induced tumor processes, while many of them are aberrantly expressed. Here, we focus on the role of Cdc42 in cell morphogenesis, proliferation, motility, angiogenesis and survival, introduce the Cdc42-targeted non-coding RNAs, as well as present current development of effective Cdc42-targeted inhibitors in breast cancer.
Collapse
|
10
|
Huang QY, Lai XN, Qian XL, Lv LC, Li J, Duan J, Xiao XH, Xiong LX. Cdc42: A Novel Regulator of Insulin Secretion and Diabetes-Associated Diseases. Int J Mol Sci 2019; 20:ijms20010179. [PMID: 30621321 PMCID: PMC6337499 DOI: 10.3390/ijms20010179] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 12/26/2018] [Accepted: 12/29/2018] [Indexed: 02/07/2023] Open
Abstract
Cdc42, a member of the Rho GTPases family, is involved in the regulation of several cellular functions including cell cycle progression, survival, transcription, actin cytoskeleton organization and membrane trafficking. Diabetes is a chronic and metabolic disease, characterized as glycometabolism disorder induced by insulin deficiency related to β cell dysfunction and peripheral insulin resistance (IR). Diabetes could cause many complications including diabetic nephropathy (DN), diabetic retinopathy and diabetic foot. Furthermore, hyperglycemia can promote tumor progression and increase the risk of malignant cancers. In this review, we summarized the regulation of Cdc42 in insulin secretion and diabetes-associated diseases. Organized researches indicate that Cdc42 is a crucial member during the progression of diabetes, and Cdc42 not only participates in the process of insulin synthesis but also regulates the insulin granule mobilization and cell membrane exocytosis via activating a series of downstream factors. Besides, several studies have demonstrated Cdc42 as participating in the pathogenesis of IR and DN and even contributing to promote cancer cell proliferation, survival, invasion, migration, and metastasis under hyperglycemia. Through the current review, we hope to cast light on the mechanism of Cdc42 in diabetes and associated diseases and provide new ideas for clinical diagnosis, treatment, and prevention.
Collapse
Affiliation(s)
- Qi-Yuan Huang
- Department of Pathophysiology, Medical College, Nanchang University, Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, 461 Bayi Road, Nanchang 330006, China.
| | - Xing-Ning Lai
- Department of Pathophysiology, Medical College, Nanchang University, Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, 461 Bayi Road, Nanchang 330006, China.
| | - Xian-Ling Qian
- Department of Pathophysiology, Medical College, Nanchang University, Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, 461 Bayi Road, Nanchang 330006, China.
| | - Lin-Chen Lv
- Department of Pathophysiology, Medical College, Nanchang University, Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, 461 Bayi Road, Nanchang 330006, China.
| | - Jun Li
- Department of Pathophysiology, Medical College, Nanchang University, Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, 461 Bayi Road, Nanchang 330006, China.
| | - Jing Duan
- Department of Pathophysiology, Medical College, Nanchang University, Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, 461 Bayi Road, Nanchang 330006, China.
| | - Xing-Hua Xiao
- Department of Pathophysiology, Medical College, Nanchang University, Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, 461 Bayi Road, Nanchang 330006, China.
| | - Li-Xia Xiong
- Department of Pathophysiology, Medical College, Nanchang University, Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, 461 Bayi Road, Nanchang 330006, China.
| |
Collapse
|
11
|
Follistatin Expression in Human Invasive Breast Tumors: Pathologic and Clinical Associations. Appl Immunohistochem Mol Morphol 2018; 26:108-112. [PMID: 27389553 DOI: 10.1097/pai.0000000000000385] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Follistatin is a potent native activin antagonist that is expressed in the normal mammary gland and in different breast proliferative diseases. Despite experimental evidence that follistatin can modulate the breast cancer cell cycle, the clinical significance of follistatin expression in these tumors is unknown. The aim of this study was to correlate the intensity of follistatin expression in invasive breast cancer with some of its clinical and pathologic features, such as the disease stage and the hormonal receptor status. Paraffin blocks of tumor samples that had been fixed in buffered formalin were obtained from 154 women subjected to surgery for breast cancer between 2008 and 2012. Sections from all paraffin blocks were cut and processed together by immunohistochemistry using a commercial monoclonal antibody to human follistatin. The intensity of follistatin staining was unrelated to the menopausal status, the disease stage, the grade, progesterone receptor expression, and local or systemic recurrence. However, follistatin immunoreactivity was significantly stronger in estrogen receptor (ER)-negative tumors than in ER-positive tumors. These findings suggest that follistatin expression in invasive breast cancer is unrelated to the disease severity and the risk of recurrence, but is more intense in ER-negative tumors.
Collapse
|
12
|
Athreya AP, Gaglio AJ, Cairns J, Kalari KR, Weinshilboum RM, Wang L, Kalbarczyk ZT, Iyer RK. Machine Learning Helps Identify New Drug Mechanisms in Triple-Negative Breast Cancer. IEEE Trans Nanobioscience 2018; 17:251-259. [PMID: 29994716 DOI: 10.1109/tnb.2018.2851997] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This paper demonstrates the ability of mach- ine learning approaches to identify a few genes among the 23,398 genes of the human genome to experiment on in the laboratory to establish new drug mechanisms. As a case study, this paper uses MDA-MB-231 breast cancer single-cells treated with the antidiabetic drug metformin. We show that mixture-model-based unsupervised methods with validation from hierarchical clustering can identify single-cell subpopulations (clusters). These clusters are characterized by a small set of genes (1% of the genome) that have significant differential expression across the clusters and are also highly correlated with pathways with anticancer effects driven by metformin. Among the identified small set of genes associated with reduced breast cancer incidence, laboratory experiments on one of the genes, CDC42, showed that its downregulation by metformin inhibited cancer cell migration and proliferation, thus validating the ability of machine learning approaches to identify biologically relevant candidates for laboratory experiments. Given the large size of the human genome and limitations in cost and skilled resources, the broader impact of this work in identifying a small set of differentially expressed genes after drug treatment lies in augmenting the drug-disease knowledge of pharmacogenomics experts in laboratory investigations, which could help establish novel biological mechanisms associated with drug response in diseases beyond breast cancer.
Collapse
|
13
|
Mohan S, Patel S, Greenstein I, Ng C, Frazier K, Nguyen G, Harding L, Barlow D. Metabolic relevance for N-hydroxy L-arginine reduction in estrogen-negative breast cancer cells. Amino Acids 2018; 50:1629-1636. [PMID: 29922922 DOI: 10.1007/s00726-018-2603-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/09/2018] [Indexed: 02/06/2023]
Abstract
We had shown Nw-hydroxy-L-arginine (NOHA) as a promising blood-based biomarker for estrogen-receptor-negative (ER-) breast cancer (BC) that differentiates ER- BC based on grade and molecular phenotype. In this in vitro study, we assessed the metabolic relevance for ER- BC-specific NOHA modulation and correlated them with NOHA regulatory responses. This study aids future NOHA clinical utility in ER- BC diagnosis and therapy management and would prove useful for potential drug discovery and development process.
Collapse
Affiliation(s)
- Srinidi Mohan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New England, Portland, ME, 04103, USA.
| | - Seema Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New England, Portland, ME, 04103, USA
| | - Ian Greenstein
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New England, Portland, ME, 04103, USA
| | - Cathy Ng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New England, Portland, ME, 04103, USA
| | - Kelly Frazier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New England, Portland, ME, 04103, USA
| | - Giang Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New England, Portland, ME, 04103, USA
| | - Lisa Harding
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New England, Portland, ME, 04103, USA
| | - David Barlow
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New England, Portland, ME, 04103, USA
| |
Collapse
|
14
|
Wei DH, Wang Y, Shi HR. Association of p53 and mitochondrial gene with chemosensitization by metformin in ovarian cancer. Oncotarget 2018; 9:2971-2976. [PMID: 29423021 PMCID: PMC5790438 DOI: 10.18632/oncotarget.22863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 11/03/2017] [Indexed: 12/04/2022] Open
Abstract
OBJECTIVE This study aims to investigate the association of p53 and D-loop gene with drug resistance and sensitization induced by metformin in ovarian cancer. RESULTS Metformin suppresses cells in a time-dependent manner, but the inhibition does not change with the dose of metformin. This suggests that within a certain range of concentration in the body, metformin has a constant inhibitory effect on cells; and the long-term use of metformin yields a better effect. CONCLUSIONS Metformin enhances the sensitivity of drug-resistant ovarian cancer cells to chemotherapy. METHODS The third passage cells of 17 cancerous ovarian tissues, which were successfully passaged five times, were used as the study objects; and the SKOV3 cell line was used as the positive control. After three adaptations, cells were cultured for 72 hours in an orthogonal experiment using drugs that were used for adaptation. Then, the inhibitory rate on cells in the experimental group was observed by CCK8 assay, in order to study the sensitization effect of metformin in different chemotherapies of ovarian cancer.
Collapse
Affiliation(s)
- De-Hua Wei
- Department of Gynecology, Puyang People's Hospital Affiliated to Xinxiang Medical University, Henan 457000, China
| | - Yan Wang
- Department of Gynecology, Puyang People's Hospital Affiliated to Xinxiang Medical University, Henan 457000, China
| | - Hui-Rong Shi
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
15
|
Thakur KK, Bordoloi D, Kunnumakkara AB. Alarming Burden of Triple-Negative Breast Cancer in India. Clin Breast Cancer 2017; 18:e393-e399. [PMID: 28801156 DOI: 10.1016/j.clbc.2017.07.013] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/16/2017] [Indexed: 01/11/2023]
Abstract
Breast cancer is the most prevalent cancer among women worldwide. Among the different breast cancer subtypes, triple-negative breast cancer (TNBC), which is more prevalent among younger age women, is the most aggressive form. Numerous clinicopathologic studies performed throughout the world strongly support the utterly poor prognoses and high recurrence rate of TNBC. The present report details a thorough data survey from Google and PubMed on the burden of TNBC worldwide and other associated factors, with special emphasis on its ever increasing incidence among Indian women. Our analysis revealed that the proportion of TNBC ranges from 6.7% to 27.9% in different countries, with the highest reported percentage in India among all, followed by Indonesia, Algeria, and Pakistan. Most of the other countries (Netherlands, Italy, London, Germany) had a TNBC incidence less than the mean level (ie, 15%). The high incidence of TNBC in the Indian population is associated with vivid risk factors, which primarily include lifestyle, deprivation status, obesity, family history, high mitotic indexes, and BRCA1 mutations. The treatment of TNBC is greatly hampered due to the lack of targeted therapies. Hence, it requires earnest attention towards extensive research for the prevention and development of treatment modalities with high efficacy.
Collapse
Affiliation(s)
- Krishan K Thakur
- Cancer Biology Laboratory & DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| | - Devivasha Bordoloi
- Cancer Biology Laboratory & DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory & DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, India.
| |
Collapse
|
16
|
Zhang X, Harbeck N, Jeschke U, Doisneau-Sixou S. Influence of vitamin D signaling on hormone receptor status and HER2 expression in breast cancer. J Cancer Res Clin Oncol 2017; 143:1107-1122. [PMID: 28025696 DOI: 10.1007/s00432-016-2325-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/16/2016] [Indexed: 12/14/2022]
Abstract
PURPOSE Breast cancer is a significant global public health issue. It is the leading cause of death among women around the world, with an incidence increasing annually. In recent years, there has been more and more information in the literature regarding a protective role of vitamin D in cancer. Increasingly preclinical and clinical studies suggest that vitamin D optimal levels can reduce the risk of breast cancer development and regulate cancer-related pathways. METHOD In this review, we focus on the importance of vitamin D in breast cancers, discussing especially the influence of vitamin D signaling on estrogen receptor and human epidermal growth factor receptor 2 (HER2), two major biomarkers of breast cancer today. CONCLUSION We discuss the possibility of actual and future targeted therapeutic approaches for vitamin D signaling in breast cancer.
Collapse
Affiliation(s)
- Xi Zhang
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany
| | - Nadia Harbeck
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany
| | - Udo Jeschke
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany
| | - Sophie Doisneau-Sixou
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany.
- Faculté des Sciences Pharmaceutiques, Université Paul Sabatier Toulouse III, 31062, Toulouse Cedex 09, France.
| |
Collapse
|
17
|
Athreya AP, Kalari KR, Cairns J, Gaglio AJ, Wills QF, Niu N, Weinshilboum R, Iyer RK, Wang L. Model-based unsupervised learning informs metformin-induced cell-migration inhibition through an AMPK-independent mechanism in breast cancer. Oncotarget 2017; 8:27199-27215. [PMID: 28423712 PMCID: PMC5432329 DOI: 10.18632/oncotarget.16109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 02/18/2017] [Indexed: 11/25/2022] Open
Abstract
We demonstrate that model-based unsupervised learning can uniquely discriminate single-cell subpopulations by their gene expression distributions, which in turn allow us to identify specific genes for focused functional studies. This method was applied to MDA-MB-231 breast cancer cells treated with the antidiabetic drug metformin, which is being repurposed for treatment of triple-negative breast cancer. Unsupervised learning identified a cluster of metformin-treated cells characterized by a significant suppression of 230 genes (p-value < 2E-16). This analysis corroborates known studies of metformin action: a) pathway analysis indicated known mechanisms related to metformin action, including the citric acid (TCA) cycle, oxidative phosphorylation, and mitochondrial dysfunction (p-value < 1E-9); b) 70% of these 230 genes were functionally implicated in metformin response; c) among remaining lesser functionally-studied genes for metformin-response was CDC42, down-regulated in breast cancer treated with metformin. However, CDC42's mechanisms in metformin response remained unclear. Our functional studies showed that CDC42 was involved in metformin-induced inhibition of cell proliferation and cell migration mediated through an AMPK-independent mechanism. Our results points to 230 genes that might serve as metformin response signatures, which needs to be tested in patients treated with metformin and, further investigation of CDC42 and AMPK-independence's role in metformin's anticancer mechanisms.
Collapse
Affiliation(s)
- Arjun P. Athreya
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Krishna R. Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Junmei Cairns
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Alan J. Gaglio
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Quin F. Wills
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Nifang Niu
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Richard Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Ravishankar K. Iyer
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
18
|
Muccio DD, Atigadda VR, Brouillette WJ, Bland KI, Krontiras H, Grubbs CJ. Translation of a Tissue-Selective Rexinoid, UAB30, to the Clinic for Breast Cancer Prevention. Curr Top Med Chem 2017; 17:676-695. [PMID: 27320329 PMCID: PMC9904082 DOI: 10.2174/1568026616666160617093604] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 03/25/2016] [Accepted: 03/25/2016] [Indexed: 11/22/2022]
Abstract
This review focuses on our efforts to translate a low-toxicity retinoid X receptor-selective agonist, UAB30, to the clinic for the prevention of breast cancers. The review is divided into several sections. First, the current status of breast cancer prevention is discussed. Next, preclinical studies are presented that support translation of rexinoids to the clinic for cancer prevention. While current FDAapproved retinoids and rexinoids demonstrate profound effects in treating cancers, they lack sufficient safety for long term use in the high risk population that is otherwise disease free. The review stresses the need to identify cancer preventive drugs that are effective and safe in order to gain wide use in the clinic. Due to the heterogeneity of the disease, UAB30 is evaluated for the prevention of ER-positive and ER-negative mammary cancers. Since selective estrogen receptor modulators and aromatase inhibitors are used clinically to prevent and treat ER-positive breast cancers, preclinical studies also must demonstrate efficacy of UAB30 in combination with existing drugs under use in the clinic. To support an Investigational New Drug Application to the FDA, data on pharmacology and toxicity as well as mutagenicity is gathered prior to human trials. The review concludes with a discussion of the outcomes of human Phase 0/1 clinical trials that determine the safety and pharmacology of UAB30. These studies are essential before this agent is evaluated for efficacy in phase 2 trials. Success in phase 2 evaluation is critical before long-term and costly phase 3 trials are undertaken. The lack of surrogate biomarkers as endpoints for phase 2 evaluation of rexinoid preventive agents is discussed.
Collapse
Affiliation(s)
- Donald D. Muccio
- Department of Chemistry, University of Alabama at Birmingham, Birmingham Alabama 35294 USA
| | - Venkatram R Atigadda
- Department of Chemistry, University of Alabama at Birmingham, Birmingham Alabama 35294 USA
| | - Wayne J Brouillette
- Department of Chemistry, University of Alabama at Birmingham, Birmingham Alabama 35294 USA
| | - Kirby I Bland
- Department of Surgery, University of Alabama at Birmingham, Birmingham Alabama 35294 USA
| | - Helen Krontiras
- Department of Surgery, University of Alabama at Birmingham, Birmingham Alabama 35294 USA
| | - Clinton J Grubbs
- Department of Surgery, University of Alabama at Birmingham, Birmingham Alabama 35294 USA
| |
Collapse
|
19
|
Afghahi A, Telli ML, Kurian AW. Genetics of triple-negative breast cancer: Implications for patient care. Curr Probl Cancer 2016; 40:130-140. [PMID: 28340968 DOI: 10.1016/j.currproblcancer.2016.09.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 09/20/2016] [Indexed: 12/13/2022]
Abstract
Patients with triple-negative breast cancer (TNBC), defined as lacking expression of the estrogen and progesterone receptors (ER/PR) and amplification of the HER2 oncogene, often have a more aggressive disease course than do patients with hormone receptor-positive breast cancer, including higher rates of visceral and central nervous system metastases, early cancer recurrences and deaths. Triple-negative breast cancer is associated with a young age at diagnosis and both African and Ashkenazi Jewish ancestry, the latter due to three common founder mutations in the highly penetrant cancer susceptibility genes BRCA1 and BRCA2 (BRCA1/2). In the past decade, there has been a surge both in genetic testing technology and in patient access to such testing. Advances in genetic testing have enabled more rapid and less expensive commercial sequencing than could be imagined only a few years ago. Massively parallel, next-generation sequencing allows the simultaneous analysis of many different genes. Studies of TNBC patients in the current era have revealed associations of TNBC with mutations in several moderate penetrance breast cancer susceptibility genes, including PALB2, BARD1, BRIP1, RAD51C and RAD51D. Interestingly, many of these genes, like BRCA1/2, are involved in homologous recombination DNA double-stranded repair. In this review, we summarize the current understanding of pathogenic germline gene mutations associated with TNBC and the early detection and prevention strategies for women at risk of developing this high-risk breast cancer subtype. Furthermore, we discuss recent the advances in targeted therapies for TNBC patients with a hereditary predisposition, including the role of poly (ADP-ribose) polymerase (PARP) inhibitors in BRCA1/2 mutation-associated breast cancers.
Collapse
Affiliation(s)
- Anosheh Afghahi
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora, CO
| | - Melinda L Telli
- Stanford University School of Medicine, Department of Medicine, Stanford, CA
| | - Allison W Kurian
- Stanford University School of Medicine, Department of Medicine, Stanford, CA; Stanford University School of Medicine, Department of Health Research and Policy, Stanford, CA.
| |
Collapse
|
20
|
Bouchard G, Therriault H, Geha S, Bérubé-Lauzière Y, Bujold R, Saucier C, Paquette B. Stimulation of triple negative breast cancer cell migration and metastases formation is prevented by chloroquine in a pre-irradiated mouse model. BMC Cancer 2016; 16:361. [PMID: 27282478 PMCID: PMC4901430 DOI: 10.1186/s12885-016-2393-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 06/01/2016] [Indexed: 12/04/2022] Open
Abstract
Background Some triple negative breast cancer (TNBC) patients are at higher risk of recurrence in the first three years after treatment. This rapid relapse has been suggested to be associated with inflammatory mediators induced by radiation in healthy tissues that stimulate cancer cell migration and metastasis formation. In this study, the ability of chloroquine (CQ) to inhibit radiation-stimulated development of metastasis was assessed. Methods The capacity of CQ to prevent radiation-enhancement of cancer cell invasion was assessed in vitro with the TNBC cell lines D2A1, 4T1 and MDA-MB-231 and the non-TNBC cell lines MC7-L1, and MCF-7. In Balb/c mice, a single mammary gland was irradiated with four daily doses of 6 Gy. After the last irradiation, irradiated and control mammary glands were implanted with D2A1 cells. Mice were treated with CQ (vehicle, 40 or 60 mg/kg) 3 h before each irradiation and then every 72 h for 3 weeks. Migration of D2A1 cells in the mammary gland, the number of circulating tumor cells and lung metastasis were quantified, and also the expression of some inflammatory mediators. Results Irradiated fibroblasts have increased the invasiveness of the TNBC cell lines only, a stimulation that was prevented by CQ. On the other hand, invasiveness of the non-TNBC cell lines, which was not enhanced by irradiated fibroblasts, was also not significantly modified by CQ. In Balb/c mice, treatment with CQ prevented the stimulation of D2A1 TNBC cell migration in the pre-irradiated mammary gland, and reduced the number of circulating tumor cells and lung metastases. This protective effect of CQ was associated with a reduced expression of the inflammatory mediators interleukin-1β, interleukin-6, and cyclooxygenase-2, while the levels of matrix metalloproteinases-2 and −9 were not modified. CQ also promoted a blocking of autophagy. Conclusion CQ prevented radiation-enhancement of TNBC cell invasion and reduced the number of lung metastases in a mouse model. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2393-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gina Bouchard
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, 3001, 12e Avenue Nord, Sherbrooke, Québec, J1H 5 N4, Canada
| | - Hélène Therriault
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, 3001, 12e Avenue Nord, Sherbrooke, Québec, J1H 5 N4, Canada
| | - Sameh Geha
- Department of Pathology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Canada
| | - Yves Bérubé-Lauzière
- Department of Electrical and Computer Engineering, Centre d'imagerie moléculaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Rachel Bujold
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, 3001, 12e Avenue Nord, Sherbrooke, Québec, J1H 5 N4, Canada.,Service of Radiation Oncology, Université de Sherbrooke, Sherbrooke, Canada
| | - Caroline Saucier
- Department of Anatomy and Cellular Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Benoit Paquette
- Centre for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, 3001, 12e Avenue Nord, Sherbrooke, Québec, J1H 5 N4, Canada.
| |
Collapse
|
21
|
Curcumin and Resveratrol as Promising Natural Remedies with Nanomedicine Approach for the Effective Treatment of Triple Negative Breast Cancer. JOURNAL OF ONCOLOGY 2016; 2016:9750785. [PMID: 27242900 PMCID: PMC4875984 DOI: 10.1155/2016/9750785] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/23/2016] [Accepted: 04/06/2016] [Indexed: 01/04/2023]
Abstract
Researchers have made considerable progress in last few decades in understanding mechanisms underlying pathogenesis of breast cancer, its phenotypes, its molecular and genetic changes, its physiology, and its prognosis. This has allowed us to identify specific targets and design appropriate chemical entities for effective treatment of most breast cancer phenotypes, resulting in increased patient survivability. Unfortunately, these strategies have been largely ineffective in the treatment of triple negative breast cancer (TNBC). Hormonal receptors lacking render the conventional breast cancer drugs redundant, forcing scientists to identify novel targets for treatment of TNBC. Two natural compounds, curcumin and resveratrol, have been widely reported to have anticancer properties. In vitro and in vivo studies show promising results, though their effectiveness in clinical settings has been less than satisfactory, owing to their feeble pharmacokinetics. Here we discuss these naturally occurring compounds, their mechanism as anticancer agents, their shortcomings in translational research, and possible methodology to improve their pharmacokinetics/pharmacodynamics with advanced drug delivery systems.
Collapse
|
22
|
Qin Y, Sundaram S, Essaid L, Chen X, Miller SM, Yan F, Darr DB, Galanko JA, Montgomery SA, Major MB, Johnson GL, Troester MA, Makowski L. Weight loss reduces basal-like breast cancer through kinome reprogramming. Cancer Cell Int 2016; 16:26. [PMID: 27042159 PMCID: PMC4818517 DOI: 10.1186/s12935-016-0300-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 03/22/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity is associated with an aggressive subtype of breast cancer called basal-like breast cancer (BBC). BBC has no targeted therapies, making the need for mechanistic insight urgent. Reducing adiposity in adulthood can lower incidence of BBC in humans. Thus, this study investigated whether a dietary intervention to reduce adiposity prior to tumor onset would reverse HFD-induced BBC. METHODS Adult C3(1)-Tag mice were fed a low or high fat diet (LFD, HFD), and an obese group initially exposed to HFD was then switched to LFD to induce weight loss. A subset of mice was sacrificed prior to average tumor latency to examine unaffected mammary gland. Latency, tumor burden and progression was evaluated for effect of diet exposure. Physiologic, histology and proteomic analysis was undertaken to determine mechanisms regulating obesity and weight loss in BBC risk. Statistical analysis included Kaplan-Meier and log rank analysis to investigate latency. Student's t tests or ANOVA compared variables. RESULTS Mice that lost weight displayed significantly delayed latency compared to mice fed HFD, with latency matching those on LFD. Plasma leptin concentrations significantly increased with adiposity, were reduced to control levels with weight loss, and negatively correlated with tumor latency. HFD increased atypical ductal hyperplasia and ductal carcinoma in situ in mammary gland isolated prior to mean latency-a phenomenon that was lost in mice induced to lose weight. Importantly, kinome analysis revealed that weight loss reversed HFD-upregulated activity of PKC-α, PKD1, PKA, and MEK3 and increased AMPKα activity in unaffected mammary glands isolated prior to tumor latency. CONCLUSIONS Weight loss prior to tumor onset protected against the effects of HFD on latency and pre-neoplastic lesions including atypical ductal hyperplasia and DCIS. Using innovative kinomics, multiple kinases upstream of MAPK/P38α were demonstrated to be activated by HFD-induced weight gain and reversed with weight loss, providing novel targets in obesity-associated BBC. Thus, the HFD-exposed microenvironment that promoted early tumor onset was reprogrammed by weight loss and the restoration of a lean phenotype. Our work contributes to an understanding of underlying mechanisms associated with tumor and normal mammary changes that occur with weight loss.
Collapse
Affiliation(s)
- Yuanyuan Qin
- />CB 7461, Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 2203 McGavran Greenberg Hall, Chapel Hill, NC 27599-7461 USA
| | - Sneha Sundaram
- />CB 7461, Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 2203 McGavran Greenberg Hall, Chapel Hill, NC 27599-7461 USA
| | - Luma Essaid
- />CB 7461, Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 2203 McGavran Greenberg Hall, Chapel Hill, NC 27599-7461 USA
| | - Xin Chen
- />Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Samantha M. Miller
- />Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Feng Yan
- />Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - David B. Darr
- />Mouse Phase I Unit, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Joseph A. Galanko
- />Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Stephanie A. Montgomery
- />Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Michael B. Major
- />Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Gary L. Johnson
- />Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
- />Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Melissa A. Troester
- />Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
- />Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Liza Makowski
- />CB 7461, Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 2203 McGavran Greenberg Hall, Chapel Hill, NC 27599-7461 USA
- />Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| |
Collapse
|
23
|
Harvie M, Howell A, Evans DG. Can diet and lifestyle prevent breast cancer: what is the evidence? Am Soc Clin Oncol Educ Book 2016:e66-73. [PMID: 25993238 DOI: 10.14694/edbook_am.2015.35.e66] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Breast cancer is the most common cancer among women in both developed and less-developed countries. Rates of breast cancer are increasing worldwide, with a particular increase in postmenopausal and estrogen receptor-positive cases. The World Cancer Research Fund (WCRF) and American Cancer Society (ACS) cancer prevention guidelines recommend maintaining a healthy weight, undertaking at least 150 minutes of moderate intensity exercise per week, limiting alcohol consumption, and eating a plant-based diet. Observational data link adherence to physical activity and alcohol guidelines throughout life to a reduced risk of developing pre- and postmenopausal breast cancer. Weight control throughout life appears to prevent cases after menopause. Adherence to a healthy dietary pattern does not have specific effects on breast cancer risk but remains important as it reduces the risk for other common diseases, such as cardiovascular disease (CVD), diabetes, and dementia. Emerging data suggest that smoking during adolescence or early adulthood increases later risk of breast cancer. Lifestyle factors appear to modify risk among high-risk women with a family history and those with typical risk of the general population, although their effects among carriers of BRCA mutations are not well defined. Recent expert reports estimate that successful lifestyle changes could prevent 25% to 30% of cases of breast cancer. These reductions will only be achieved if we can implement targeted prevention programs for high-risk women and women in population-based breast screening programs during childhood, adolescence, and early adulthood when the rapidly developing breast is particularly susceptible to carcinogenesis.
Collapse
Affiliation(s)
- Michelle Harvie
- From the Genesis Breast Cancer Prevention Centre, University Hospital of South Manchester NHS Foundation Trust, Manchester, United Kingdom
| | - Anthony Howell
- From the Genesis Breast Cancer Prevention Centre, University Hospital of South Manchester NHS Foundation Trust, Manchester, United Kingdom
| | - D Gareth Evans
- From the Genesis Breast Cancer Prevention Centre, University Hospital of South Manchester NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
24
|
Abstract
Breast cancer is a highly heterogeneous disease. Tamoxifen is a selective estrogen receptor (ER) modulator and is mainly indicated for the treatment of breast cancer in postmenopausal women and postsurgery neoadjuvant therapy in ER-positive breast cancers. Interestingly, 5–10% of the ER-negative breast cancers have also shown sensitivity to tamoxifen treatment. The involvement of molecular markers and/or signaling pathways independent of ER signaling has been implicated in tamoxifen sensitivity in the ER-negative subgroup. Studies reveal that variation in the expression of estrogen-related receptor alpha, ER subtype beta, tumor microenvironment, and epigenetics affects tamoxifen sensitivity. This review discusses the background of the research on the action of tamoxifen that may inspire future studies to explore effective therapeutic strategies for the treatment of ER-negative and triple-negative breast cancers, the latter being an aggressive disease with worse clinical outcome.
Collapse
Affiliation(s)
- Subrata Manna
- Department of Biology, Yeshiva University, New York, NY, USA
| | - Marina K Holz
- Department of Biology, Yeshiva University, New York, NY, USA; Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
25
|
Lee O, Khan SA. Novel routes for administering chemoprevention: local transdermal therapy to the breasts. Semin Oncol 2016; 43:107-115. [DOI: 10.1053/j.seminoncol.2015.09.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
26
|
De Hert M, Peuskens J, Sabbe T, Mitchell AJ, Stubbs B, Neven P, Wildiers H, Detraux J. Relationship between prolactin, breast cancer risk, and antipsychotics in patients with schizophrenia: a critical review. Acta Psychiatr Scand 2016; 133:5-22. [PMID: 26114737 DOI: 10.1111/acps.12459] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/02/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE A recent meta-analysis showed that breast cancer probably is more common in female patients with schizophrenia than in the general population (effect size = 1.25, P < 0.05). Increasing experimental and epidemiological data have alerted researchers to the influence of prolactin (PRL) in mammary carcinogenesis. We therefore investigated the possible relationship between antipsychotic-induced hyperprolactinemia (HPRL) and breast cancer risk in female patients with schizophrenia. METHOD A literature search (1950 until January 2015), using the MEDLINE database, was conducted for English-language published clinical trials to identify and synthesize data of the current state of knowledge concerning breast cancer risk (factors) in women with schizophrenia and its (their) relationship between HPRL and antipsychotic medication. RESULTS Although an increasing body of evidence supports the involvement of PRL in breast carcinogenesis, results of human prospective studies are limited, equivocal, and correlative (with risk ratios ranging from 0.70 to 1.9 for premenopausal women and from 0.76 to 2.03 for postmenopausal women). Moreover, these studies equally do not take into account the local production of PRL in breast epithelium, although amplification or overexpression of the local autocrine/paracrine PRL loop may be a more important mechanism in tumorigenesis. Until now, there is also no conclusive evidence that antipsychotic medication can increase the risk of breast malignancy and mortality. CONCLUSION Other breast risk factors than PRL, such as nulliparity, obesity, diabetes mellitus, and unhealthy lifestyle behaviours (alcohol dependence, smoking, low physical activity), probably are of greater relevance in individual breast cancer cases within the population of female patients with schizophrenia.
Collapse
Affiliation(s)
- M De Hert
- Department of Neurosciences, KU Leuven University Psychiatric Centre, Kortenberg, Belgium
| | - J Peuskens
- Department of Neurosciences, KU Leuven University Psychiatric Centre, Kortenberg, Belgium
| | - T Sabbe
- Department of Neurosciences, KU Leuven University Psychiatric Centre, Kortenberg, Belgium
| | - A J Mitchell
- Department of Psycho-oncology, Cancer & Molecular Medicine, University of Leicester, Leicester, UK
| | - B Stubbs
- School of Health and Social Care, University of Greenwich, Greenwich, UK
| | - P Neven
- Multidisciplinary Breast Center, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium
| | - H Wildiers
- Multidisciplinary Breast Center, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium.,Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium
| | - J Detraux
- Department of Neurosciences, KU Leuven University Psychiatric Centre, Kortenberg, Belgium
| |
Collapse
|
27
|
Grassi D, Ghorbanpoor S, Acaz-Fonseca E, Ruiz-Palmero I, Garcia-Segura LM. The Selective Estrogen Receptor Modulator Raloxifene Regulates Arginine-Vasopressin Gene Expression in Human Female Neuroblastoma Cells Through G Protein-Coupled Estrogen Receptor and ERK Signaling. Endocrinology 2015. [PMID: 26200092 DOI: 10.1210/en.2014-2010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The selective estrogen receptor modulator raloxifene reduces blood pressure in hypertensive postmenopausal women. In the present study we have explored whether raloxifene regulates gene expression of arginine vasopressin (AVP), which is involved in the pathogenesis of hypertension. The effect of raloxifene was assessed in human female SH-SY5Y neuroblastoma cells, which have been recently identified as a suitable cellular model to study the estrogenic regulation of AVP. Raloxifene, within a concentration ranging from 10(-10) M to 10(-6) M, decreased the mRNA levels of AVP in SH-SY5Y cells with maximal effect at 10(-7) M. This effect of raloxifene was imitated by an agonist (±)-1-[(3aR*,4S*,9bS*)-4-(6-bromo-1,3-benzodioxol-5-yl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinolin-8-yl]-ethanone of G protein-coupled estrogen receptor-1 (GPER) and blocked by an antagonist (3aS*,4R*,9bR*)-4-(6-bromo-1,3-benzodioxol-5-yl)-3a,4,5,9b-3H-cyclopenta[c]quinoline of GPER and by GPER silencing. Raloxifene induced a time-dependent increase in the level of phosphorylated ERK1 and ERK2, by a mechanism blocked by the GPER antagonist. The treatment of SH-SY5Y cells with either a MAPK/ERK kinase 1/2-specific inhibitor (1,4-diamino-2, 3-dicyano-1,4-bis(2-aminophenylthio)butadine) or a protein kinase C inhibitor (sotrastaurin) blocked the effects of raloxifene on the phosphorylation of ERK1/2 and the regulation of AVP mRNA levels. These results reveal a mechanism mediating the regulation of AVP expression by raloxifene, involving the activation of GPER, which in turn activates protein kinase C, MAPK/ERK kinase, and ERK. The regulation of AVP by raloxifene and GPER may have implications for the treatment of blood hypertension(.).
Collapse
Affiliation(s)
- Daniela Grassi
- Instituto Cajal (D.G., S.G., E.A.-F., I.R.P., L.M.G.-S.), Consejo Superior de Investigaciones Científicas, E-28002 Madrid, Spain; Institute of Anatomy and Cell Biology (D.G.), Department of Molecular Embryology, Albert-Ludwigs-Universität Freiburg, D-79104 Freiburg, Germany; and Department of Cell and Molecular Biology (S.G.), School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Samar Ghorbanpoor
- Instituto Cajal (D.G., S.G., E.A.-F., I.R.P., L.M.G.-S.), Consejo Superior de Investigaciones Científicas, E-28002 Madrid, Spain; Institute of Anatomy and Cell Biology (D.G.), Department of Molecular Embryology, Albert-Ludwigs-Universität Freiburg, D-79104 Freiburg, Germany; and Department of Cell and Molecular Biology (S.G.), School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Estefania Acaz-Fonseca
- Instituto Cajal (D.G., S.G., E.A.-F., I.R.P., L.M.G.-S.), Consejo Superior de Investigaciones Científicas, E-28002 Madrid, Spain; Institute of Anatomy and Cell Biology (D.G.), Department of Molecular Embryology, Albert-Ludwigs-Universität Freiburg, D-79104 Freiburg, Germany; and Department of Cell and Molecular Biology (S.G.), School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Isabel Ruiz-Palmero
- Instituto Cajal (D.G., S.G., E.A.-F., I.R.P., L.M.G.-S.), Consejo Superior de Investigaciones Científicas, E-28002 Madrid, Spain; Institute of Anatomy and Cell Biology (D.G.), Department of Molecular Embryology, Albert-Ludwigs-Universität Freiburg, D-79104 Freiburg, Germany; and Department of Cell and Molecular Biology (S.G.), School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Luis M Garcia-Segura
- Instituto Cajal (D.G., S.G., E.A.-F., I.R.P., L.M.G.-S.), Consejo Superior de Investigaciones Científicas, E-28002 Madrid, Spain; Institute of Anatomy and Cell Biology (D.G.), Department of Molecular Embryology, Albert-Ludwigs-Universität Freiburg, D-79104 Freiburg, Germany; and Department of Cell and Molecular Biology (S.G.), School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| |
Collapse
|
28
|
Hatoum D, McGowan EM. Recent advances in the use of metformin: can treating diabetes prevent breast cancer? BIOMED RESEARCH INTERNATIONAL 2015; 2015:548436. [PMID: 25866793 PMCID: PMC4383151 DOI: 10.1155/2015/548436] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/21/2014] [Indexed: 12/16/2022]
Abstract
There is substantial epidemiological evidence pointing to an increased incidence of breast cancer and morbidity in obese, prediabetic, and diabetic patients. In vitro studies strongly support metformin, a diabetic medication, in breast cancer therapy. Although metformin has been heralded as an exciting new breast cancer treatment, the principal consideration is whether metformin can be used as a generic treatment for all breast cancer types. Importantly, will metformin be useful as an inexpensive therapy for patients with comorbidity of diabetes and breast cancer? In general, meta-analyses of clinical trial data from retrospective studies in which metformin treatment has been used for patients with diabetes and breast cancer have a positive trend; nevertheless, the supporting clinical data outcomes remain inconclusive. The heterogeneity of breast cancer, confounded by comorbidity of disease in the elderly population, makes it difficult to determine the actual benefits of metformin therapy. Despite the questionable evidence available from observational clinical studies and meta-analyses, randomized phases I-III clinical trials are ongoing to test the efficacy of metformin for breast cancer. This special issue review will focus on recent research, highlighting in vitro research and retrospective observational clinical studies and current clinical trials on metformin action in breast cancer.
Collapse
Affiliation(s)
- Diana Hatoum
- School of Medical and Molecular Biosciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Eileen M. McGowan
- School of Medical and Molecular Biosciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
- School of Medicine, University of Sydney, Camperdown, Sydney, NSW 2006, Australia
| |
Collapse
|
29
|
Abstract
Breast cancer is the most common cancer of women in Western Europe and North America. Effective strategies of medical prevention could reduce the burden of breast cancer mortality. The best evidence for a risk reduction exists for hormonal agents such as tamoxifen and raloxifene (22-72%) or aromatase inhibitors (50-65%). However, the severity of side effects and the lack of evidence for an improved survival compromise the risk/benefit balance. In this review the results of chemoprevention studies, including new treatment approaches, are summarized with critical discussion of their use in clinical practice.
Collapse
Affiliation(s)
- Johannes Stubert
- Department of Obstetrics and Gynecology, University of Rostock, Germany
| | - Max Dieterich
- Department of Obstetrics and Gynecology, University of Rostock, Germany
| | - Bernd Gerber
- Department of Obstetrics and Gynecology, University of Rostock, Germany
| |
Collapse
|
30
|
Chu NJ, Armstrong TD, Jaffee EM. Nonviral oncogenic antigens and the inflammatory signals driving early cancer development as targets for cancer immunoprevention. Clin Cancer Res 2015; 21:1549-57. [PMID: 25623216 DOI: 10.1158/1078-0432.ccr-14-1186] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 12/18/2014] [Indexed: 12/13/2022]
Abstract
Cancer immunoprevention is an emerging field that holds much promise. Within the past 20 years, prophylactic vaccines have been implemented on the population level for the immunoprevention of carcinomas induced by viruses, specifically hepatitis B virus (HBV) and human papillomavirus (HPV) infection. Armed with the success of prophylactic vaccines that prevent viral-induced tumors, the field must overcome its next hurdle: to develop robust prophylactic vaccines that prevent the remaining >80% of human cancers not induced by viral infection. In this review, we discuss some of the most promising non-virus-associated prophylactic vaccines that target endogenous neoantigens, including the earliest oncogene products, altered mucin 1 (MUC1) and α-enolase (ENO1), all of which produce new targets in the earliest stages of nonviral-induced tumorigenesis. We also highlight a novel attenuated Listeria monocytogenes-based vaccine expressing mutant oncogene Kras(G12D) (LM-Kras) effective in a pancreatic cancer model. A novel chimeric human/rat HER-2 plasmid vaccine (HuRT-DNA vaccine) effective in a breast cancer model is also discussed. In addition to prophylactic vaccine developments, this review highlights the potential use of classic drugs, such as aspirin and metformin, as chemopreventive agents that can potentially be used as adjuvants to enhance the anticancer immunogenicity and efficacy of noninfectious prophylactic vaccines by modulating the inflammatory pathways within the early tumor microenvironment (TME) that propels tumorigenesis. Finally, timing of prophylactic vaccine administration is critical to its immunopreventive efficacy, providing a necessary role of current and emerging biomarkers for cancer screening and early cancer detection.
Collapse
Affiliation(s)
- Nina J Chu
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Todd D Armstrong
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth M Jaffee
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|