Breast Cancer: Current Trends in Molecular Research -  - E-Book

Breast Cancer: Current Trends in Molecular Research E-Book

0,0
79,78 €

-100%
Sammeln Sie Punkte in unserem Gutscheinprogramm und kaufen Sie E-Books und Hörbücher mit bis zu 100% Rabatt.
Mehr erfahren.
Beschreibung

Breast cancer is one of the most common cancer types worldwide, and is a leading cause of cancer related deaths in women. In this book, medical experts review our current understanding of the molecular biology and characteristics of breast cancer. The topics covered in this book provide comprehensive knowledge of mechanisms underlying breast carcinogenesis, and are intended for a wide audience including scientists, teachers, and students.

11 chapters present information about several topics on breast cancer, including the role of cell growth and proliferation pathways, androgen and cytokine signaling, germline mutations in breast cancer susceptibility genes, and molecular factors causing invasive and metastatic breast cancer. In addition, the editors discuss the recent advancements in multi-omics data analysis based on inter-and intra-tumor molecular profiles.

The reference highlights how the knowledge and understanding of the biological behavior of breast neoplasms have facilitated ongoing investigations into dietary polyphenolic compounds with antioxidant properties, making them function as cancer chemopreventive agents. Along with this, the current development of treatment strategies such as targeted molecular therapy, and radiation therapy is brought to the fore to update readers.

Sie lesen das E-Book in den Legimi-Apps auf:

Android
iOS
von Legimi
zertifizierten E-Readern

Seitenzahl: 538

Veröffentlichungsjahr: 2002

Bewertungen
0,0
0
0
0
0
0
Mehr Informationen
Mehr Informationen
Legimi prüft nicht, ob Rezensionen von Nutzern stammen, die den betreffenden Titel tatsächlich gekauft oder gelesen/gehört haben. Wir entfernen aber gefälschte Rezensionen.



Table of Contents
BENTHAM SCIENCE PUBLISHERS LTD.
End User License Agreement (for non-institutional, personal use)
Usage Rules:
Disclaimer:
Limitation of Liability:
General:
FOREWORD
PREFACE
List of Contributors
Cellular and Molecular Mechanisms of Breast Cancer Progression
Abstract
INTRODUCTION
TYPES OF BREAST CANCER
Histological Subtypes
Non-invasive (or in situ) Breast Cancer
Invasive or Infiltrating Breast Cancer
Metastatic Breast Cancer
Molecular Subtypes
Luminal A
Luminal B
Triple-Negative/basal-like
HER2-enriched
Normal-like
CELLULAR AND MOLECULAR BASIS OF BREAST CANCER PROGRESSION
Molecular Mechanisms of Breast Cancer
Cellular Mechanisms of Breast Cancer Progression
Immune Cells and Breast Cancer Progression
Innate Immunity in Breast Cancer
NK Cells
Myeloid-derived Suppressor Cells (MDSCs)
Macrophages
Adaptive Immunity in Breast Cancer
T Regulatory Cells (Tregs)
B-lymphocytes
T-lymphocytes
SUMMARY AND FUTURE DIRECTIONS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Immune-Endocrine Perspectives of Breast Cancer
Abstract
INTRODUCTION
ORIGIN AND EVOLUTION OF BREAST CANCER
ANDROGEN RECEPTOR (AR) AND BREAST CANCER
Androgen Receptor in the Normal Breast
AR in Breast Cancer
AR as a Prognostic Factor for Breast Cancer
AR as a Therapeutic Target for Breast Cancer
CANCER CELLS AND IMMUNE RESPONSE
MECHANISM OF CANCER ESCAPE FROM IMMUNE SURVEILLANCE
ALTERING IMMUNOGENIC CHARACTERISTICS
SUPPRESSION OF IMMUNE RESPONSE BY IMMUNOSUPPRESSIVE FACTORS
Immunosuppressive Cells
Outpacing the Immune System
Cytokines in Breast Cancer
Cytokines and Steroids Synthesis
ANDROGENS AND THE IMMUNE SYSTEM
Androgens and Innate Immune System
Neutrophils
Macrophages (Mφ)
Dendritic Cells (Dcs)
Natural Killer Cells (NK Cells)
ANDROGENS AND ADAPTIVE IMMUNITY
ANDROGENS AND CYTOKINES
CONCLUDING REMARKS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
DNA Damage Response: A Therapeutic Landscape for Breast Cancer Treatment
Abstract
INTRODUCTION
Risk Factors
SUBTYPES
Histological Classification
Molecular Classification
GENETIC PREDISPOSITION
GENOMIC INSTABILITY, DNA DAMAGE RESPONSE (DDR), AND CANCER
DNA AS A TARGET FOR CANCER THERAPY
Genomic Overview of Breast Cancer
Targeting DNA and DDR Response Associated Factors for Breast Cancer Therapy
NOVEL THERAPEUTIC STRATEGIES TARGETING DDR ASSOCIATED PATHWAYS FOR BREAST CANCER TREATMENT
PARP Inhibitors (PARPi)
DNA-PK Inhibitor (DNAPKi)
FEN1 Inhibitor
ATR/CHK1 Inhibitor
Wee1 Inhibitor
FUTURE DIRECTIONS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Emerging Trends in Bioinformatics for Breast Cancer Molecular Research
Abstract
INTRODUCTION
MULTI-OMICS DATA IN BREAST CANCER RESEARCH
Molecular Subtyping of Breast Cancer Using Multi-omics Data
PAN-CANCER STUDY USING MULTI-OMICS DATA
BIOINFORMATICS METHODS IN BREAST CANCER RESEARCH
BIOINFORMATICS FOR UNDERSTANDING BREAST CANCER RESPONSE
BIOINFORMATICS TOOLS AND TECHNIQUES
CONCLUDING REMARKS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Role of Nitric Oxide in Breast Cancer
Abstract
INTRODUCTION
Physiological and Biological Action of Nitric Oxide
Various Roles of Nitric Oxide in Promoting Carcinogenesis
INVOLVEMENT OF NITRIC OXIDE IN VARIOUS TYPES OF CANCERS
Nitric Oxide in Cervix Uteri Cancer
HPV (Human Papillomavirus), Nitric Oxide, and Cervix Carcinoma
Nitric Oxide in Gastric Carcinoma
Nitric Oxide, H. pylori, and Gastric Carcinoma
Nitric Oxide in Head and Neck Carcinoma
Human Papillomavirus, Nitric Oxide, and Head Neck Carcinoma
Nitric Oxide and Lung Carcinoma
Nitric Oxide in Brain Tumor
Breast Cancer and Nitric Oxide
NITRIC OXIDE AND HORMONES INVOLVED IN BREAST CANCER
TUMORICIDAL VERSUS THE TUMOR-PROMOTING EFFECT OF Nitric Oxide
NITRIC OXIDE AS A BREAST CANCER THERAPEUTIC AGENT
CONCLUSIONS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Autoantibodies as Clinical Biomarkers in Breast Cancer
Abstract
INTRODUCTION
Biomarkers
Breast Cancer Biomarkers
AUTOANTIBODIES
GOALS OF CANCER BIOMARKERS
AUTOANTIBODIES AS POTENTIAL CLINICAL MARKERS FOR CANCER
TECHNIQUES FOR AUTOANTIBODIES IDENTIFICATION
Recombinant cDNA Expression Cloning or SEREX
Serological Proteome Analysis or SERPA
Protein Microarray and Multiple Affinity Protein Profiling Or MAPPING
Protein Microarray
AUTOANTIBODIES TO INDIVIDUAL TUMOR ANTIGENS IN BREAST CANCER
AUTOANTIBODY PANELS FOR EARLY DETECTION OF BREAST CANCERS
CONCLUSIONS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Epigenetics of Breast Cancer
Abstract
INTRODUCTION
Epigenetic Programming and Cellular Physiology
Epigenetic Modifications are Strongly Interlinked
Epigenetic Imbalance in Cancer
BREAST CANCER AND EPIGENETICS
Methylome of Breast Cancer
Histone Modification in BC
EPIGENETICS IN CANCER PREVENTION AND THERAPY
EPIGENETICS AS A BIOMARKER OF CANCER
EPIGENETIC THERAPY IN CANCER
DNMT Inhibitors
HDAC Inhibitors
COMBINATION THERAPY
CONCLUDING REMARKS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGMENTS
REFERENCES
Nanoparticles Targeting and Uptake: Current Advances in Breast Cancer Research
Abstract
INTRODUCTION
INTERNALIZATION PATHWAYS
PHAGOCYTOSIS
PINOCYTOSIS
Clathrin-Mediated Endocytosis
Caveolae Mediated Endocytosis
Clathrin-Caveolae Independent Endocytosis
Macropinocytosis
RECEPTOR-MEDIATED INTERNALIZATION OF NANO-MEDICINE
Epidermal Growth Factor Receptor
CD44 Receptor
Folate Receptor (FR)
Formyl Peptide Receptor
LAT1 Transporter
Transferrin Receptor (TFR)
α/β Integrins
ESCAPING PHAGOCYTIC CLEARANCE
CONCLUSIONS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Dietary Polyphenols and its Molecular Mechanism in the Management of Breast Cancer
Abstract
INTRODUCTION
Breast Cancer: An Overview
Molecular Subtypes of Breast Cancer
MOLECULAR MECHANISM OF ACTION OF POLYPHENOLS IN BREAST CANCER
Polyphenolic Compounds and Redox Balance
Polyphenolic Compounds and Uncontrolled Proliferation
Polyphenolic Compounds Regulating Apoptosis
Polyphenolic Compounds and Modulation of Inflammation-Related Factors
Polyphenolic Compounds and Modulation of the Estrogen Receptor
CONCLUSIONS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Radiotherapy in Carcinoma Breast
Abstract
INTRODUCTION
RADIOTHERAPY: DEFINITION AND PRINCIPLE
RADIATION SOURCES IN EXTERNAL BEAM RADIOTHERAPY
Telegamma (CO-60) Unit
MEDICAL LINEAR ACCELERATOR
Stand
Gantry
Head
Couch
Control Console
CARCINOMA BREAST
BREAST CANCER: TREATMENT MODALITIES OTHER THAN RADIOTHERAPY
EXTERNAL BEAM RADIOTHERAPY IN CA BREAST: DIAGNOSIS AND STAGING
STEPS IN EXTERNAL BEAM RADIOTHERAPY
SIMULATION
CT (VIRTUAL) SIMULATION: CLINICAL SETTINGS
TREATMENT PLANNING
CONTOURING WORKSTATION
Practical Aspects: Sites for Irradiation, Volume Concept
Postmastectomy Radiotherapy
Whole Breast Radiotherapy
Accelerated Partial Breast Irradiation
Regional Nodal Irradiation
TREATMENT PLANNING WORKSTATION: DOSE CALCULATION
CONCEPT OF DOSE FRACTIONATION
Conventional Radiotherapy
Hypofractionation
Hyper Fractionation
THREE-DIMENSIONAL CONFORMAL RADIOTHERAPY (3DCRT)
DCRT With Uniform (Constant) Photon Fluence
INTENSITY MODULATED RADIOTHERAPY
DOSE AND FRACTIONATION OF RADIOTHERAPY
TREATMENT PLAN EVALUATION
VERIFICATION AND RADIATION DELIVERY
BIOMARKERS (Prognostic and Predictive) in Breast Cancer Management
CONCLUDING REMARKS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
An Overview of Breast Cancer Therapy
Abstract
INTRODUCTION
Neo-Adjuvant Therapy and Adjuvant Therapy
Surgery
Radiation Therapy
Chemotherapy
Hormone Therapy
Immunotherapy
SUMMARY AND CONCLUDING REMARKS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Breast Cancer: Current Trends in Molecular Research
Edited By
Shankar Suman
Comprehensive Cancer Center
The Ohio State University
Columbus
USA
Shivam Priya
Comprehensive Cancer Center
The Ohio State University
Columbus
USA
&
Akanksha Nigam
Department of Microbiology and Molecular Genetics
IMRIC, the Hebrew University- Hadassah Medical School
Jerusalem
Israel

BENTHAM SCIENCE PUBLISHERS LTD.

End User License Agreement (for non-institutional, personal use)

This is an agreement between you and Bentham Science Publishers Ltd. Please read this License Agreement carefully before using the ebook/echapter/ejournal (“Work”). Your use of the Work constitutes your agreement to the terms and conditions set forth in this License Agreement. If you do not agree to these terms and conditions then you should not use the Work.

Bentham Science Publishers agrees to grant you a non-exclusive, non-transferable limited license to use the Work subject to and in accordance with the following terms and conditions. This License Agreement is for non-library, personal use only. For a library / institutional / multi user license in respect of the Work, please contact: [email protected].

Usage Rules:

All rights reserved: The Work is 1. the subject of copyright and Bentham Science Publishers either owns the Work (and the copyright in it) or is licensed to distribute the Work. You shall not copy, reproduce, modify, remove, delete, augment, add to, publish, transmit, sell, resell, create derivative works from, or in any way exploit the Work or make the Work available for others to do any of the same, in any form or by any means, in whole or in part, in each case without the prior written permission of Bentham Science Publishers, unless stated otherwise in this License Agreement.You may download a copy of the Work on one occasion to one personal computer (including tablet, laptop, desktop, or other such devices). You may make one back-up copy of the Work to avoid losing it.The unauthorised use or distribution of copyrighted or other proprietary content is illegal and could subject you to liability for substantial money damages. You will be liable for any damage resulting from your misuse of the Work or any violation of this License Agreement, including any infringement by you of copyrights or proprietary rights.

Disclaimer:

Bentham Science Publishers does not guarantee that the information in the Work is error-free, or warrant that it will meet your requirements or that access to the Work will be uninterrupted or error-free. The Work is provided "as is" without warranty of any kind, either express or implied or statutory, including, without limitation, implied warranties of merchantability and fitness for a particular purpose. The entire risk as to the results and performance of the Work is assumed by you. No responsibility is assumed by Bentham Science Publishers, its staff, editors and/or authors for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products instruction, advertisements or ideas contained in the Work.

Limitation of Liability:

In no event will Bentham Science Publishers, its staff, editors and/or authors, be liable for any damages, including, without limitation, special, incidental and/or consequential damages and/or damages for lost data and/or profits arising out of (whether directly or indirectly) the use or inability to use the Work. The entire liability of Bentham Science Publishers shall be limited to the amount actually paid by you for the Work.

General:

Any dispute or claim arising out of or in connection with this License Agreement or the Work (including non-contractual disputes or claims) will be governed by and construed in accordance with the laws of the U.A.E. as applied in the Emirate of Dubai. Each party agrees that the courts of the Emirate of Dubai shall have exclusive jurisdiction to settle any dispute or claim arising out of or in connection with this License Agreement or the Work (including non-contractual disputes or claims).Your rights under this License Agreement will automatically terminate without notice and without the need for a court order if at any point you breach any terms of this License Agreement. In no event will any delay or failure by Bentham Science Publishers in enforcing your compliance with this License Agreement constitute a waiver of any of its rights.You acknowledge that you have read this License Agreement, and agree to be bound by its terms and conditions. To the extent that any other terms and conditions presented on any website of Bentham Science Publishers conflict with, or are inconsistent with, the terms and conditions set out in this License Agreement, you acknowledge that the terms and conditions set out in this License Agreement shall prevail.

Bentham Science Publishers Ltd. Executive Suite Y - 2 PO Box 7917, Saif Zone Sharjah, U.A.E. Email: [email protected]

FOREWORD

Breast cancer malignancy is now becoming a global leading cause of cancer related death among females all around the world. The current developments in breast cancer research have wrought to increase the life expectancy in patients in the 21st century but a long way to go to cure this deadly disease. Among the major challenges, the heterogeneity in cancer cells make the disease more complex. Researchers have made a significant advancement in studying the heterogeneous features in breast cancer and multiple subsets of breast cancer are discovered. In the molecular biology studies, breast cancer stem-like cells, driver mutations and changes in tumor microenvironment are investigated as potentially hallmarks for the disease progression. Immunological aspects in breast cancer are considered cutting-edge science in recent discoveries. I am happy to look at this book that has incorporated all current advances in the breast cancer research. I congratulate Drs. Suman and Priya for editing a nice compilation of the different pieces of findings of breast cancer research in this book. I hope this will help to understand the recent advances in breast cancer to the researchers’ particularly new investigators and clinicians.

Chandan Singh, PhD Assistant Professor, Department of Biochemistry Banaras Hindu University India

PREFACE

Breast cancer is the most frequent malignancy in women worldwide. It is associated with several risk factors including mutations, inheritance, and environmental factors. The heterogeneity in breast cancer cells shows the complexity of this disease. Despite newer day-to-day developments in cancer research, advanced breast cancer is still challenging to manage. The scientific innovation in breast cancer research has led to an increase in patient's overall survival in the last few decades. For example- the discovery of sensitive screening tools enhanced early-stage detection of breast cancer and further novel treatment regimens improved therapeutic benefits. The current research is also focused on developing new surgical modalities that are minimally invasive, and new radiation modalities with minimal side effects. This book is intended to target a broad audience who has an interest in breast cancer research and therapy. We have incorporated chapters delineating the recent studies on breast cancer with an emphasis on etiology, diagnosis, and therapy. Chapters uncover the new updated information on breast cancer signaling, immune-response, DNA damage, and epigenetic modifications with bioinformatics resources. The book will allow readers to review the data of numerous studies on immunotherapy and gene therapy as well. The latest concept on the use of nanotechnology in breast cancer, has also been reviewed in detail as nanotechnology has opened a paradigm shift in targeted drug delivery in breast cancer. The specific chapter also shows the importance of dietary polyphenol and its role in breast cancer. The application of radiotherapy in breast cancer is also well described, which will be helpful to gain the concept of clinical radiotherapy. In overall, chapters are organized to give the readers a practical and efficient way to familiarize themselves with the newest ongoing research in the field of breast cancer.

Shankar Suman Comprehensive Cancer Center The Ohio State University Columbus USAShivam Priya Comprehensive Cancer Center The Ohio State University Columbus USA &Akanksha Nigam Department of Microbiology and Molecular Genetics

List of Contributors

Ajeet Kumar Verma840 Biomedical Research Tower, Wexner Medical Centre, The Ohio State University, Columbus Ohio, USAAkhtar ParwezBhuwaneshwari Dayal College (B.D. College), Patna, IndiaAlkhansa S. MahmoudDepartment of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, University Putra Malaysia, 43400 Serdang, Selango, Malaysia Radiobiology Department, Sudan Atomic Energy Commission, 1111 Khartoum, SudanAlok Kumar PandeyNanomaterial Toxicology Laboratory, Nanomaterial Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, IndiaArchana LalwaniDepartment of Botany and biotechnology, Sadhu Vaswani Autonomous College, Sant Hirdaram Nagar, Bhopal, 462030, IndiaChandra Bhushan PrasadDepartment of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, IndiaDeepika SinghDepartment of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, IndiaEkhlaque A. KhanDepartment of Biotechnology, Chaudhary Bansi Lal University, Bhiwani, Haryana, IndiaGirish RaiVivekanand Government PG College Maiha, MP, India Centre of Biomedical Research, SGPGIMS Campus, Lucknow, UP, IndiaHazilawati H.J. HamzahDepartment of Veterinary Pathology and Microbiology, Faculty Veterinary Medicine, University Putra Malaysia, 43400 Serdang, Selangor, MalaysiaKaran Singh SainiGovt. Kamla Nehru Girls College, Balaghat, M.P., IndiaM.N. Mohd HezmeeDepartment of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, University Putra Malaysia, 43400 Serdang, Selango, MalaysiaManuraj PandeyDepartment of Biotechnology, Unique College, Jawahar Chowk, T.T. Nagar, Bhopal, M.P., IndiaMranalini VermaDepartment of Radiotherapy, King George’s Medical University, UP Lucknow, IndiaOnila LugunNanomaterial Toxicology Laboratory, Nanomaterial Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, IndiaPuja Rani MinaDivision of Gastroenterology, Department of Internal Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USAPrachi GuptaDepartment of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, Wisconsin, USARajendra MehtaDepartment of Rural Technology and Social Development, Guru Ghasidas Vishwavidyalaya (Central University), Bilaspur, CG, IndiaRatnasekhar ChCSIR-Central Institute of Medicinal and Aromatic Plants Lucknow, Lucknow, IndiaRituraj KonwarCSIR- Central Drug Research Institute, Lucknow, IndiaSammed N. MandapeCenter for Human Identification, University of North Texas Health Science Center, Fort Worth, Texas, USASanjay Mishra840 Biomedical Research Tower, Wexner Medical Centre, The Ohio State University, Columbus Ohio, USASarfraj Ahmad SiddiquiDepartment of Zoolog, University of Lucknow, Lucknow, IndiaShankar SumanBiomedical Research Tower, Comprehensive Cancer Center, Ohio, USASwati Misri840 Biomedical Research Tower, Wexner Medical Centre, The Ohio State University, Columbus Ohio, USATeerthraj VermaDepartment of Radiotherapy, King George’s Medical University, UP Lucknow, IndiaTengku Ahbriza F.T.A.Malaysian Nuclear Agency, Ministry of Science, Technology and Innovation, Bangi, 43000 Kajang, SelangorZuki AB. ZakariaDepartment of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, University Putra Malaysia, 43400 Serdang, Selango, Malaysia

Cellular and Molecular Mechanisms of Breast Cancer Progression

Ajeet Kumar Verma1,*,Sanjay Mishra1,Puja Rani Mina2,Swati Misri1
1 840 Biomedical Research Tower, Wexner Medical Centre, The Ohio State University, Columbus Ohio, USA
2 Division of Gastroenterology, Department of Internal Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA

Abstract

Breast cancer is a common death-related cancer in women globally. Early and non-metastatic stage breast cancers are curable in 70-80% of the patients, while advanced-stage distant organ metastatic breast cancers are incurable with present treatment options. Although multiple risk factors are associated with breast cancer, among them, genetic predispositions in BRCA1 and BRCA2 genes are the most causative factor for breast cancer malignancy. The initiation and progression of breast cancer is a multi-step process, which can initiate either in ducts or lobules of the breast tissues. As time progresses pre-invasive lesions form of breast neoplasm transforms into atypical ductal hyperplasia (ADH), ductal carcinoma in situ (DCIS)/lobular carcinoma in situ (LCIS), and eventually become invasive carcinoma. The molecular mechanisms behind the initiation and progression of breast cancer are not completely understood. However, epithelial-mesenchymal transition (EMT) is the assurance of malignancy which disrupts endothelial integrity and therefore, it increases the spreading of cancer cells and facilitates metastasis. After the epithelial-mesenchymal transition of tumor cells, tumor cells invade and migrate the neighboring as well as distant tissues, cross the endothelial barrier and enter the blood, and attach to a secondary site, forming metastases. In this chapter, we have reviewed an overview of the molecular mechanisms of breast cancer progression.

Keywords: EMT, Hyperplasia, Malignancy, Treg, Tumor cells.
*Corresponding author Ajeet Kumar Verma: 840 Biomedical Research Tower, 460 West 12th Avenue, Columbus, Ohio, USA-43210; E-mail: [email protected]

INTRODUCTION

Breast cancer is the most common carcinoma and has a high incidence rate amongst all types of women cancer worldwide [1]. Breast cancer is a heterogeneous disease, which can be divided into several subtypes based on histological appearance and the expression of molecular markers.

In the United States, it is approximated that one in eight women will develop breast cancer in her lifetime. This development of breast cancer is a complex process, which is multistep events from initiation, progression, to metastasis. The mechanistic basis of breast cancer metastasis is the epithelial to mesenchymal transition (EMT) [2].

Cancer cell migratory nature is defined by EMT and its converse MET (mesenchymal-epithelial transition) process. There are a series of events that happen when cells transform from epithelial to mesenchymal stages, like cell junction loss and acquiring the migrant nature. During the transformation process, the cells lose their “epithelial” characteristics such as cell to cell adhesion, cell junctions, and cells that express vimentin as the key intermediate filament protein. Undoubtedly, EMT is a shorthand that means changes in the cell shape from coherent “epithelial” monolayer to a migratory fibroblastic or “mesenchymal” phenotype.

The discovery of more driver genes and the complex molecular pathways of breast cancer pave a better understanding of disease progression. Based on the expression profiles of various genes, breast cancer has been categorized into five distinct subtypes: luminal A, luminal B, normal-like, basal-like, and human epithelial growth factor receptor 2 (HER2) types. All these subtypes have different clinical consequences and therapeutic options [3]. The oncogenes strongly influence the changes in malignancy and distant metastasis. Normal breast stromal cells transform into cancer cells by gain-in-function mutations (oncogene). These mutant genes are driver oncogenes that dysregulate apoptotic pathways to become resistance phenotypes. Further constant oncogenic pressure dilutes the effect of existing chemotherapies and thereby leads to poor patient survival. Thus, targeting oncogenic drivers and their downstream signaling molecules are being pursued rationally for breast cancer. For example, luminal or HER2-positive subtypes of breast cancers are getting treated with endocrine therapies or HER2 targeted therapies. In addition, molecular mechanism-based therapies have brought a paradigm shift in recent therapy. Such therapies consist of DNA repair PARP protein inhibitors for BRCA-mutant basal cancer subtype or CDK4/6 inhibitors for advanced ER+ HER2- breast cancers. Several clinical trials are uncovering the potential therapeutic use of immune checkpoint inhibitors as monotherapy or with other target-based therapies for breast cancer. Additionally, the role of different immune cells and molecular markers in the development and metastasis of breast cancer has been explored in past decades which led to the development and use of immunotherapy for breast cancer patients.

The family history of having breast cancer or if any close relatives, such as a mother, sister, or daughter ever been diagnosed with breast cancer, the probability of having breast cancer rises at an early stage i.e., premenopausal age [4-6]. Despite the advancements in treatment, the five-year survival risks for patients with metastatic breast cancer are still only very low (22%) [7], and breast cancer still directly affects one in every eight U.S. women [8]. Breast cancers can be sub-categorized based on the expression of distinct molecular and histological markers. Infiltrating ductal carcinoma (~85%) and infiltrating lobular carcinoma (~15%) are categorized as the main histological subtypes of invasive breast cancer [6]. Approximately 75% of all Breast cancer patients have molecular signatures that are designated as hormone receptor-positive cancers, expressing either estrogen receptor (ER) or progesterone receptor (PR) at higher than 1%. Because of the receptor's overexpression, these cancers can be targeted by drugs that directly target the receptors like tamoxifen or aromatase inhibitors. Another group of treatable breast cancers includes human epidermal growth factor receptor 2 (HER2) positive (15-20%), which tend to grow faster than HER2 negative breast cancers, but it can be targeted with anti-Her2 therapies such as trastuzumab [6]. While there has been a success in finding drugs that treat and cure early-stage, ER, PR, and HER2 positive breast cancers, there are no approved target-based therapies to date for triple-negative breast cancer (TNBC). The TNBC patients (~15%) do not display high amounts of any of these molecular markers [6]. For this reason, patients with TNBC have a higher likelihood of recurrence and lower five-year survival rates than those diagnosed with other subtypes of breast cancer.

TYPES OF BREAST CANCER

Breast cancers are classified based on their presence in different areas of the breast such as lobules, ducts, or within tissues. However, based on cell origin, breast cancers are broadly categorized as carcinomas and sarcomas. Carcinomas are the type of breast cancer that arises from epithelial components lying between lobules and terminal ducts. Sarcomas are a very rare form of breast cancer (it is less than 1% of total breast cancer) that arises from the stromal constituents of the breast, these include myofibroblasts and blood vessel cells. These categorizations are inadequate because sometimes a single mammary tumor can be a mixture of different cell types [9-11]. Breast cancers generally fall into two subtypes, histological subtypes, and molecular subtypes.

Histological Subtypes

Most breast cancers are diagnosed with carcinoma. Under carcinomas, many different types of breast cancer are recognized based on invasiveness compared to the primary tumor sites. Breast cancers fall under three major groups based on pathological characteristics and invasive properties which are non-invasive (or in situ), invasive, and metastatic breast cancer types [9-11].

Non-invasive (or in situ) Breast Cancer

DCIS or ductal carcinoma in situ is a type of breast cancer that starts initiating in the milk duct and has not spread to the remaining part of the breast tissue. DCIS is a pre-or non-invasive type of breast cancer that is also well known as intraductal carcinoma. Although DCIS is a non-invasive, it has a high possibility to be invasive cancer, so early, efficient and suitable treatment is necessary for the patients to inhibit invasive cancer development [12-14].

Invasive or Infiltrating Breast Cancer

The term invasive (or infiltrating) breast cancer defines breast cancer cells that spread (invade) into the neighboring breast tissue. Invasive breast cancers are further divided into two categories, invasive ductal carcinoma (IDC) and invasive lobular carcinoma (ILC). Invasive breast cancer has cancer cells that invade and spread outside of the normal breast lobules and ducts. Approximately two-thirds of the women diagnosed with invasive breast cancer are older than 55 years. Invasive breast cancers have the potential to spread to lymph nodes and other body organs from the original site. Invasive ductal carcinoma is the most common type and constitutes about 80% of all breast cancers. IDC is further divided into tubular, medullary, papillary, mucinous, and cribriform carcinoma of the breast. While ILC is the second most common type and accounts for 10-15% of all breast cancers. Although ILC is the most frequent among older women, it can also affect women at an early age [12-14].

Metastatic Breast Cancer

Metastatic breast cancers are advanced, late-stage types of breast cancer that have spread to other body organs like lymph nodes, liver, lung, brain, and bone. About 30% of the women diagnosed with early-stage breast cancer develop metastatic breast cancer. Breast cancer metastasis depends on the unique molecular and cell biology of the tumor and the stage at the time of the initial diagnosis [12-14].

Molecular Subtypes

Classification of breast cancer based on molecular components is more useful than the classification based on histology for the treatment planning and development of newer targeted therapies. There are five molecular subtypes of breast cancer, luminal A, luminal B, triple-negative or basal-like, HER2-enriched, and normal-like Table 1.

Table 1Molecular subtypes of breast cancers.Molecular SubtypesReceptor StatusCharacteristicsLuminal AER+, PR±, HER2-, Low Ki67~70%, Most common best prognosisLuminal BER+, PR±, HER2±, High Ki6710%–20%, Lower survival than Luminal ATriple-negative/Basal-likeER-, PR-, HER2-15%–20%, More common in black women, Diagnosed at a younger age, Worst prognosisHER2-enrichedER-, PR-, HER2+5%–15%Normal-likeER+, PR±, HER2-, Low Ki67Rare, Low proliferation gene cluster expression

Luminal A

This subtype is slow-growing, low-grade, and has the best prognosis. This subtype is hormone-receptor-positive (ER+ and/or PR+) and HER2 negative. It also expresses low levels of Ki-67 protein, which is associated with cell proliferation [15, 16].

Luminal B

This subtype is slightly fast-growing than the luminal A subtype and has a slightly worse prognosis. This subtype is hormone-receptor-positive (ER+ and/or PR+) and they are either HER2 positive or HER2 negative with high Ki67 expression levels [15, 16].

Triple-Negative/basal-like

This subtype is hormone-receptor negative (ER- and/or PR-) and HER2 negative. This subtype is most common in women with BRCA1 gene mutations. Also, this subtype is more common in younger and Afro-American women [15, 16].

HER2-enriched

This subtype is hormone-receptor negative (ER- and/or PR-) and HER2 positive. This subtype grows faster than luminal cancers and has a worse prognosis, but often can be successfully treated with targeted therapy against Her2 protein like Herceptin (trastuzumab), Tykerb (lapatinib), Nerlynx (neratinib), etc. [15, 16].

Normal-like

This subtype is like the luminal A subtype, hormone-receptor-positive (ER+ and/or PR+), HER2 negative, and expresses low levels of Ki67 protein. Although normal-like breast cancer has a good prognosis, its prognosis is slightly worse than luminal A subtype [15, 16].

CELLULAR AND MOLECULAR BASIS OF BREAST CANCER PROGRESSION

Molecular Mechanisms of Breast Cancer

Breast cancer is the accumulation of diverse malignancies that establish themselves in the mammary glands. As a result of rapid signs of advances in molecular biology, our understanding of breast cancer progression has been extended to cellular, molecular and genomic levels.

There are several cellular and molecular factors involved in breast cancer progression. Among molecular factors, integrins play a prime role in cancer cell motility and survival and interaction of cells to the extracellular matrix [17]. Matrix metalloproteinases (MMPs) belong to the zinc-dependent endopeptidases family, and they can degrade the extracellular matrix (ECM) and facilitate protein degradation at the invadopodium facing side of the invasive breast cancer cells [18, 19]. In normal epithelial tissues, E-cadherin is a cell-cell effector that plays a prime role in cancer metastasis [20, 21]. Decreased level of E-cadherin in breast cancer affects cell-cell interaction and is associated with metastatic potential. Also the downregulation of such proteins is the reason for the poor prognosis of triple-negative breast cancer patients [22, 23]. In lobular breast carcinoma, there is a loss of invasion-suppressor function due to E-cadherin gene (CDH1) mutation [24].

Epithelial to mesenchymal transition (EMT) transition is a cellular procedure playing a prime role in cancer advancement and metastasis [25]. Decreased E-cadherin is a crucial indicator of EMT transition. Reducing epithelial markers in epithelial cancer cells promotes the ability of cancer cells to invade, metastasize and release proteases that break down the extracellular matrix (ECM) [26]. Increased N-cadherin and decreased E-cadherin expression of breast cancer cells increase their ability to invade and metastasize [27]. Cancer cells undergoing EMT change their epithelial adherent type of morphology and look like mesenchymal motile cells [28]. Reduction in the epithelial cell markers expression, including E-cadherin, occludin, and cytokeratin initiates the EMT process. An increase in the mesenchymal cell markers expression, including vimentin and N-cadherin has been observed in the EMT process. Some precise transcription factors like SNAIL, SLUG, TWIST, ZEB1, and ZEB2 promote EMT by downregulating E-cadherin expression [29]. EMT transcription factors control some pathways like transforming growth factor-β (TGFβ), Wingless/β-catenin, and the phosphatidylinositol 3' kinase serine/ threonine kinase (PI3K/ AKT) which are associated with poor prognosis of breast cancer.

TGFβ also has the potential to induce EMT, it has been demonstrated that TGFβ functions as a tumor suppressor as well as a metastatic activator. At the initial stage of tumor progression, TGFβ arrests the tumor growth and induces cell apoptosis [30]. Though tumor suppressor roles of TGFβ are overpowered when the tumor progresses, and cancer cells are exposed to TGFβ. During this stage metastatic potential is enhanced due to the transition of tumor cells from epithelial phenotypes to mesenchymal phenotypes [31]. TGFβ signaling pathway functions are either SMAD-dependent or SMAD-independent pathways. TGFβ ligand binds to type I and type II serine/threonine receptors during the SMAD-dependent signaling pathway. TβRII phosphorylates TβRI and thus phosphorylated TβRI phosphorylates SMAD2 and SMAD3and activates these proteins. In the cytoplasm, SMAD2/3 interacts with SMAD4 forming a complex that is then transported into the nucleus where this complex binds with zinc finger protein (GLI1) and modulates the gene transcription [32]. Curiously, in a mammary epithelial model, SMAD2 and SMAD3 were observed to be upregulated and initiate EMT transition [33]. TGFβ signaling might also get activated independently of the SMAD signaling pathway through the PI3K/AKT pathway which regulates various cellular pathways [34]. PI3K activates AKT, which is a serine/threonine kinase that activates various downstream effectors to control cell proliferation and inhibit apoptosis hence increasing cell survival. mTOR is amongst downstream targets of the PI3K/AKT pathway which induces cell proliferation [34]. Further, AKT/mTOR activation governs glycogen synthase kinase-3β (GSK3β) and nuclear factor-ĸ B (NFĸB). GSK3β modulates many cellular responses such as cell cycle and apoptosis. Activated NFĸB stimulates and increases cell viability, cell proliferation as well as malignant transformation [35]. There is some evidence in support of the upregulated PI3K/AKT/mTOR pathway in breast cancer [36]. Additionally, SMAD-dependent and SMAD-independent pathways regulate transcription factors that are involved in EMT progressions such as TWIST, SNAIL, and SLUG [37].

Angiogenesis is the process of blood vessel formation, and the process is required for the survival, invasion, and metastasis of growing tumors. Angiogenesis has been widely studied in-depth; those working for anticancer drug discovery have focused on the anti-angiogenesis therapeutic approach [38-40]. Clinical data show that breast cancer is angiogenesis-dependent cancer [41]. Several angiogenesis-inducing factors have been identified. Among angiogenesis factors vascular endothelial growth factor (VEGF) potentially enhances angiogenesis in many types of cancer [42]. VEGF regulates vascular permeability by inducing endothelial cell proliferation which results in new vessel formation. VEGF is associated with relapse-free survival, overall survival, or both as observed in clinical studies [43]. Early-stage breast cancer patients with higher VEGF expression show a higher recurrence rate and death than low-angiogenic tumor patients [44], despite conventional adjuvant therapy treatment. VEGF binds and activates VEGFR1 and VEGFR2 receptors present on endothelial cells and induces endothelial cell motility, vascular permeability, cell survival, and proliferation [45, 46]. Although the role of VEGFR1 in cancer angiogenesis is still under investigation, the effect of this receptor on cancer angiogenesis has been recognized broadly [47-51]. VEGF signaling increases vascular permeab- ility and enables metastasis progression in cancer patients. VEGF not only triggers angiogenesis and increases breast cancer aggressiveness, but it also has many non-angiogenic functions [52]. In breast cancer cells VEGF pathway increases cell survival through AKT and extracellular signal-regulated kinase (ERK) signaling [53]. These key molecular markers help cancer cells to avoid apoptosis [54] and increase migration [44].

Cellular Mechanisms of Breast Cancer Progression

In the breast cancer tissues, some cells promote breast cancer progression, while others are opposed to breast cancer progression. This equilibrium is maintained by various cytokines and other factors secreted by immune cells. The tumor microenvironment (TME) has a tremendous contribution to breast tumor heterogeneity and cancer development, including initiation and progression of metastasis. Several cell populations are present in the tumor microenvironment which include different immune cells, tumor-associated fibroblasts, endothelial precursors, mesenchymal stromal/stem cells (MSC), adipocytes, and mature cells. Further, several other factors including cytokines, chemokines, growth factors, hormones, metabolites, and constituents of the extracellular matrix (ECM) support tumor maturation and make the tumors diverse. Interestingly, when MSC interacts with breast cancer cells, it establishes the probable carcinoma stem cell niche which generates cancer stem cell-like cells (CSCs) or tumor-initiating cells (TICs) [55-60]. The normal breast duct has luminal epithelial cells, which are covered with myoepithelial cells. These myoepithelial cells yield and attach to the basement membrane. The breast microenvironment has various stromal cell types, including fibroblasts, adipocytes, endothelial cells, immune cells, and adipocytes and it is composed of an extracellular matrix (ECM) [61]. Macrophages, myoepithelial and endothelial cells, including various ECM molecules are components of the breast microenvironment and play a prime role in mammary duct morphogenesis [62].

Breast tumors evolve through a sequential process starting from epithelial cell proliferation and progression to invasive and metastatic carcinomas [63]. During breast cancer progression, tumor-associated stromal cells have altered phenotypes and altered methylation patterns [64]. Fibroblasts support breast cancer invasion by disrupting the myoepithelial cell layer and basement membrane. Sonic hedgehog (HH) and transforming growth factor β (TGFβ) pathways are activated in myoepithelial cells which transform fibroblasts into myofibroblasts to support fibroblasts in breast cancer progression. Normal fibroblast cells produce and remodel the extracellular matrix (ECM) while carcinomas-associated fibroblasts (CAFs) have tumor-promoting roles [65]. CAFs secrete stromal-derived factor 1 (SDF1 /CXCL1), which promotes tumor cell proliferation and induces angiogenesis in a paracrine manner via CXCR4 signaling [66]. There are many hypotheses about the origin of CAFs and the widely accepted hypothesis supports that overproduced signals from neighboring tumor epithelial cells change the native phenotypes of interstitial fibroblasts and transform them into CAFs. Alternatively, bone marrow-derived mesenchymal stem cells are employed to the tumor sites by endocrine stimulation from tumor-derived factors and differentiate the native interstitial fibroblast cells into CAFs. Fibroblast cells synthesize matrix metalloproteinases (MMPs, an endopeptidase family) and degrade extracellular matrix (ECM). MMPs (MMP3 and MMP7) induce the activation of cytokines, chemokines, and various growth factors to increase tumor cell proliferation and promote tumor progression. MMPs also have a role in promoting angiogenesis (MMP1, 2, 9, and 14) [67].

Immune Cells and Breast Cancer Progression

Immune cells are one of the most important cells playing a role in breast cancer progression. However, the mechanisms of communication between immune cells and tumor cells are not well understood. Immune cells are dynamic cells present within tumors. Leukocytes might have a role in the invasiveness of breast cancer [68]. Different immune cells, such as macrophages, B cells, T cells, Cd4+ and CD8+ cytotoxic T cells infiltrate in the tumor parenchyma and tumor stroma. In the tumor microenvironment, immune cells have two different roles which are either pro-tumorigenic or anti-tumorigenic. M1 macrophages (classically activated or anti-tumorigenic), mature dendritic cells, NK cells, B cells, CD8+ T cells, CD4+ and Th1 cells, etc. participate in tumor eradication, however, M2 macrophages (alternatively activated or pro-tumorigenic), myeloid-derived suppressor cells (MDSCs), CD4+ Th2 cells, regulatory B cells, CD4+ Tregs cells, etc. are tumor promoters [69, 70]. Tumor-associated macrophages (TAMs) degrade the extracellular matrix (ECM) and induce tumor invasion. TAMs also promote angiogenesis by EGFR (epidermal growth factor receptor) signaling activation, and secretion of various proteases, and paracrine signaling between tumor cells [71].

Innate Immunity in Breast Cancer

NK Cells

NK cells are categorized as the components of the innate immune system, however, the role of NK cells in breast cancer is not clearly understood to date. Although some studies have shown that NK cells prevent and inhibit early and late metastatic cancer. NK cells do not acquire any training to fight against specific antigens. They are trained to fight against any unnaturally changed cells and this is accomplished by immunoglobulin-like MHC class I-specific receptors present on NK cell surfaces. These receptors phosphorylate internal inhibitory immunoreceptor tyrosine-based inhibitory motifs on the identification of generally expressed MHC class I on functional host cells. NK cells are the first line of protection in the context of mammalian immunity [72]. If there is any defect in NK cell-mediated cytotoxicity, it affects the initial stages of human tumorigenesis. In breast cancer, various features of host immunity are either altered or compromised, changes in the number and function of NK cells are one of them [73]. The cytokines produced by breast cancer cells can change the nature of healthy NK cells to favor metastases instead of anti-tumor activity. Some reports have shown that breast cancer cells evade NK cells by undergoing dormant stage and downregulating activating receptors [74].

Myeloid-derived Suppressor Cells (MDSCs)

The MDSCs are the distinct population of myeloid origin, which show pro-tumor activating through inhibition of cytotoxic immune cells [75]. These cells function as suppressors of T-cell responses with high arginase 1 activity. High MDSCs are a well-known indicator of poor prognostic markers for various cancers and are involved in the pro-tumorigenic pathway by inhibiting and suppressing anti-tumor responses of the host [75, 76]. Breast cancer patients have a higher number of circulating MDSCs compared to normal matched controls [77]. A higher number of MDSCs have been isolated from breast cancer patients compared to healthy control which results in a poor prognosis of breast cancer. When these cells are cultured in vitro with T cells, these cells inhibit the lymphocyte population significantly in comparison to MDSCs isolated from normal healthy controls [77]. Early-stage breast cancer patients with high neutrophil counts (a type of MDSCs) have a higher neutrophil to lymphocyte ratio and there is more chance of cancer relapse in these patients [78].

Macrophages

Macrophages play a very important physiological role in the development and function of various tissues like the brain and mammary gland. In the tumor microenvironment, macrophages are abundantly distributed inflammatory cells. Macrophages can account for up to 50 percent of the mass of breast tumors [79]. Macrophages are recruited through the expression of local chemo-attractants like colony-stimulating factor 1 (CSF1) and macrophage chemoattractant protein 1 (MCP1). Overexpression of both these chemo-attractants is correlated with the poor prognosis in various types of tumors. Macrophages are recruited to the tumor site via potent chemo-attractants and their normal functions are changed to promote tumor progression and metastasis.

MIF (Macrophage migration inhibitory factor) is a chemo-attractant and plays an important role in tumor development. Tumor cells secrete MIF, which activates metalloproteinase production by macrophages and supports tumor cell invasion [80]. Macrophages are broadly divided into anti-tumorigenic (M1) and pro-tumorigenic (M2) macrophages. Macrophages are an integral part of the innate immune system of the body. Macrophages play an important role in the protection from intracellular pathogens and cancer cells [81]. INF-γ activated M1 macrophage secrete proinflammatory cytokines for example interferons and interleukins which promote inflammation. M1 macrophages release IL-12 and IL-23 in high amounts [82]. M1 macrophages show their anti-tumor activity by acting as antigen-presenting cells [83]. They have the potential to kill intracellular pathogens by stimulating the inducible NO synthase (iNOS) gene and producing nitric oxide (NO). In this way, M1 macrophages protect the host from infections and tumor cells [84].

The M2 macrophages encourage tumor growth and metastasis, and these macrophages are stated as tumor-associated macrophages [85]. As distinct from M1 macrophages, M2 macrophages are stimulated by immune complexes or other cytokines and induce Th2 type responses [85]. Breast cancer also has a hypoxia condition, since they have a low oxygen supply. This low oxygen develops necrotic bed area in the tumor due to a sudden decrease in oxygen supply which results in cell death. Cell death debris in this hypoxic environment inside the tumor influences macrophage function and attracts them [86]. Due to hypoxia transcription factors, HIF-1α and HIF-2α are upregulated in the macrophages. HIF-1α is a hypoxia marker and it makes breast cancer most aggressive and malignant [87]. HIF-2α in TAMs increases tumor vascularity by upregulating the expression of VEGF in breast cancer [88].

Adaptive Immunity in Breast Cancer

T Regulatory Cells (Tregs)

Tregs are capable of interfering with cell mediated (Th1) and humoral responses (Th2), Treg cells play a vital role in nourishing tolerance against self-antigens and overpowering undesirable immune responses. Treg cells have a very efficient mechanism to encounter the diverse range of self/foreign peptides, the activation of these cells is regulated by higher expression of accessory molecules. In this way, Treg cells provide immune tolerance to the host by suppressing immune response through discrimination between self-and non-self-antigens. Additionally, Treg cells prevent antitumor immunity and stimulate tumor growth [89]. Several subtypes of Treg cells have been identified and characterized as CD8+, and CD4+Treg cells [89]. A transcription factor forkhead box P3 (Foxp3) is expressed by Treg cells, which is the most important Treg cell marker. Many other transcription factors, molecular chaperones, histone proteins, and RNA binding proteins bind directly or indirectly with FoxP3 in response to external stimuli [90]. Tumor cells induce Treg cells to compromise the cytotoxic immune system response to tumor antigens [91]. Treg cells express various proteins, including FOX3, IL-10, and transforming growth factor β (TGF-β) to suppress cytotoxic T cells in growing tumors in mice [92].

Upon tumor progression, breast cancer cells accumulate Fox3 positive Treg cells, transient depletion of Treg cells in prevailing, immune-suppressive breast tumors induce anti-tumor immunity in primary and metastatic tumors [93]. Many genes expressed by tumor cells are modulated by FOXP3 and it binds to the upstream of the transcription start site of CCR7 and CXCR4 genes. CCR7 and CXCR4 are chemokine receptors that are reported to be involved in cancer invasion and metastasis [94]. FoxP3 is also known to be expressed on breast cancer tumor cells and associated with a poor prognosis. Higher FoxP3 expression was observed in breast tumor tissue compared to normal breast tissue [95]. Cytokine TGF-β is secreted by Treg cells, which induces tumor growth. Antigen-presenting cells like dendritic cells activate Treg cells while it encounters tumor-associated antigen. This can lead to an immune escape, inhibition of effector T cells, and development & progression of tumor [96]. Many studies have shown the direct correlation between VEGF expression and tumor vascularity and malignancy. TGF-β1 secreted by activated Treg cells can induce VEGF expression which can increase tumor vascularity and development [97].

B-lymphocytes

B lymphocytes are well-known contributors to generating immune responses against cancer by secreting antigen-specific immunoglobulins. Early neoplastic cells are removed by acutely activated B-cells. B-cell precursors mature in the bone marrow and there is a series of immunoglobulin genes recombination that occurs to provide a varied range of B cell receptors on B-lymphocyte’s surface. Mature B-cells enter into the secondary lymphoid organs like lymph nodes and spleen. While encountering antigens, B-lymphocytes undergo clonal expansion to enhance their capacity to recognize diverse foreign antigens [98]. During breast cancer progression, breast tumor-associated stroma and secondary lymphoid organs have enriched mature B lymphocytes (native and activated B cells). Dividing/mature B-cells are enriched in draining lymph nodes of breast cancer patients compared to healthy control [99]. B-lymphocytes are present inside the breast tumor-associated stroma and play a prominent role in breast cancer. Numerous reports confirmed that infiltrating B-lymphocytes are predominant in all lymphocytes in the premalignant breast tissue and breast hyperplasia [100, 101]. Invasive breast cancers have a higher number of B cells, which account for approximately 60% of the lymphocyte population comprising neoplasia [102].

T-lymphocytes

Although B lymphocytes are predominated during early breast carcinogenesis, T-lymphocytes are associated with breast cancer progression [103]. Both CD4+ and CD8+ infiltrating T-lymphocytes are widespread in invasive and higher-grade DCIS breast cancer [104]. Approximately 1% to 45% of the total cellular mass of invasive breast carcinomas comprises infiltrated T-cells [105]. The presence of T-lymphocytes in rapidly proliferating tumors can serve as a great prognostic indicator [106]. Infiltration of a particular T-lymphocytes differs considerably, which is involved in the disease advancement and generally patient survival.

The high occurrence of CD4+ T-helper cells correlates with disease advancement, metastasis to lymph nodes, and tumor size increase [105]. It is observed that CD4+ T cells mediated cytotoxic-T lymphocyte responses can be involved in either minimizing or eradicating cancer occurrence, although CD8+ T lymphocytes are not more effective. CD8+ T lymphocytes are generally active in primary tumor sites and activated under the stimulation of various cytokines and chemokines that can be matured into Th1 cells or Th2 cells [107]. Active stimulants induce Th1-polarized CD4+ T-helper cells to secrete IFN-γ, TGF-β, TNFα, and IL-2 [108]. Together with the cytotoxic properties of CD8+ T cells, these cytokines speed up the antigen processing by the proteasomal system [109]. These cytokines stimulate MHC class I and class II expression along with antigen displaying cofactors in neoplastic cells. CD4+ Th1 cells secrete IFN-γ and stimulate the activation of macrophage cytotoxic activity by M1 macrophages [110]. In contrast, Th2-polarized CD4+ T lymphocytes release IL-4, IL-5, IL-6, IL-10, and IL-13 and prompt T-cell-derived cytotoxicity loss and promote humoral immunity (B-cell function) [111]. The infiltration of specific subtypes of T lymphocytes differs significantly and may upset disease advancement and overall patient survival. Altogether Th1 responses are favorable toward anti-tumor immunity [112, 113].

SUMMARY AND FUTURE DIRECTIONS

Breast cancer is one of the causes of cancer-related mortality in women worldwide. Breast tumor cells hijack the normal, cellular molecular signaling of the cells to achieve tumorigenesis and cell proliferation across the breast tissues. Gradually the mechanisms behind tumorigenesis and cell proliferation become better understood and DNA mutations leading to tumorigenesis are also explored. An in-depth study of the cellular and molecular mechanisms of breast cancer has the potential to lead to efficient treatment regimes.

CONSENT FOR PUBLICATION

Not applicable.

CONFLICT OF INTEREST

The authors declare no conflict of interest, financial or otherwise.

ACKNOWLEDGEMENTS

Declared none.

REFERENCES

[1]Dubey AK, Gupta U, Jain S. Breast cancer statistics and prediction methodology: a systematic review and analysis. Asian Pac J Cancer Prev APJCP 2015; 16(10): 4237-45.[http://dx.doi.org/10.7314/APJCP.2015.16.10.4237] [PMID: 26028079][2]Thiery JP, Sleeman JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol 2006; 7(2): 131-42.[http://dx.doi.org/10.1038/nrm1835] [PMID: 16493418][3]Sørlie T, Perou CM, Tibshirani R, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA 2001; 98(19): 10869-74.[http://dx.doi.org/10.1073/pnas.191367098] [PMID: 11553815][4]Ruddon RW. Cancer Biology 2007.[5]Shiovitz S, Korde LA. Genetics of breast cancer: a topic in evolution. Ann Oncol 2015; 26(7): 1291-9.[http://dx.doi.org/10.1093/annonc/mdv022] [PMID: 25605744][6]O’Sullivan CC, Loprinzi CL, Haddad TC. Updates in the Evaluation and Management of Breast Cancer. Mayo Clin Proc 2018; 93(6): 794-807.[http://dx.doi.org/10.1016/j.mayocp.2018.03.025] [PMID: 29866283][7]Kwak T, Drews-Elger K, Ergonul A, et al. Targeting of RAGE-ligand signaling impairs breast cancer cell invasion and metastasis. Oncogene 2017; 36(11): 1559-72.[http://dx.doi.org/10.1038/onc.2016.324] [PMID: 27669433][8]Schneider AP, II, Zainer CM, Kubat CK, Mullen NK, Windisch AK. The breast cancer epidemic: 10 facts. Linacre Q 2014; 81(3): 244-77.[http://dx.doi.org/10.1179/2050854914Y.0000000027] [PMID: 25249706][9]Sharma GN, Dave R, Sanadya J, Sharma P, Sharma KK. Various types and management of breast cancer: an overview. J Adv Pharm Technol Res 2010; 1(2): 109-26.[PMID: 22247839][10]Polyak K. Breast cancer: origins and evolution. J Clin Invest 2007; 117(11): 3155-63.[http://dx.doi.org/10.1172/JCI33295] [PMID: 17975657][11]Allison KH. Molecular pathology of breast cancer: what a pathologist needs to know. Am J Clin Pathol 2012; 138(6): 770-80.[http://dx.doi.org/10.1309/AJCPIV9IQ1MRQMOO] [PMID: 23161709][12]Veronesi U, Boyle P, Goldhirsch A, Orecchia R, Viale G. Breast cancer. Lancet 2005; 365(9472): 1727-41.[http://dx.doi.org/10.1016/S0140-6736(05)66546-4] [PMID: 15894099][13]Collaborative Group on Hormonal Factors in Breast CancerFamilial breast cancer: collaborative reanalysis of individual data from 52 epidemiological studies including 58,209 women with breast cancer and 101,986 women without the disease. Lancet 2001; 358(9291): 1389-99.[http://dx.doi.org/10.1016/S0140-6736(01)06524-2] [PMID: 11705483][14]Hulka BS. Epidemiology of susceptibility to breast cancer. Prog Clin Biol Res 1996; 395: 159-74.[PMID: 8895988][15]Liang Q, Ma D, Gao RF, Yu KD. Effect of Ki-67 Expression Levels and Histological Grade on Breast Cancer Early Relapse in Patients with Different Immunohistochemical-based Subtypes. Sci Rep 2020; 10(1): 7648.[http://dx.doi.org/10.1038/s41598-020-64523-1] [PMID: 32376868][16]Colditz GA, Kaphingst KA, Hankinson SE, Rosner B. Family history and risk of breast cancer: nurses’ health study. Breast Cancer Res Treat 2012; 133(3): 1097-104.[http://dx.doi.org/10.1007/s10549-012-1985-9] [PMID: 22350789][17]Crowe DL, Shuler CF. Regulation of tumor cell invasion by extracellular matrix. Histol Histopathol 1999; 14(2): 665-71.[PMID: 10212827][18]Reunanen N, Kahari V. Matrix metalloproteinases in cancer cell invasion. Madame Curie Bioscience Database 2000-2013.[19]Kelly T, Yan Y, Osborne RL, et al. Proteolysis of extracellular matrix by invadopodia facilitates human breast cancer cell invasion and is mediated by matrix metalloproteinases. Clin Exp Metastasis 1998; 16(6): 501-12.[http://dx.doi.org/10.1023/A:1006538200886] [PMID: 9872598][20]Li DM, Feng YM. Signaling mechanism of cell adhesion molecules in breast cancer metastasis: potential therapeutic targets. Breast Cancer Res Treat 2011; 128(1): 7-21.[http://dx.doi.org/10.1007/s10549-011-1499-x] [PMID: 21499686][21]Pećina-Slaus N. Tumor suppressor gene E-cadherin and its role in normal and malignant cells. Cancer Cell Int 2003; 3(1): 17.[http://dx.doi.org/10.1186/1475-2867-3-17] [PMID: 14613514][22]Wendt MK, Taylor MA, Schiemann BJ, Schiemann WP. Down-regulation of epithelial cadherin is required to initiate metastatic outgrowth of breast cancer. Mol Biol Cell 2011; 22(14): 2423-35.[http://dx.doi.org/10.1091/mbc.e11-04-0306] [PMID: 21613543][23]Li P, Sun T, Yuan Q, Pan G, Zhang J, Sun D. The expressions of NEDD9 and E-cadherin correlate with metastasis and poor prognosis in triple-negative breast cancer patients. OncoTargets Ther 2016; 9: 5751-9.[http://dx.doi.org/10.2147/OTT.S113768] [PMID: 27703373][24]Berx G, Becker KF, Höfler H, van Roy F. Mutations of the human E-cadherin (CDH1) gene. Hum Mutat 1998; 12(4): 226-37.[25]Gotzmann J, Mikula M, Eger A, et al. Molecular aspects of epithelial cell plasticity: implications for local tumor invasion and metastasis. Mutat Res 2004; 566(1): 9-20.[http://dx.doi.org/10.1016/S1383-5742(03)00033-4] [PMID: 14706509][26]Voulgari A, Pintzas A. Epithelial-mesenchymal transition in cancer metastasis: mechanisms, markers and strategies to overcome drug resistance in the clinic. Biochim Biophys Acta 2009; 1796(2): 75-90.[PMID: 19306912][27]Maret D, Gruzglin E, Sadr MS, et al. Surface expression of precursor N-cadherin promotes tumor cell invasion. Neoplasia 2010; 12(12): 1066-80.[http://dx.doi.org/10.1593/neo.10954] [PMID: 21170270][28]Yilmaz M, Christofori G. Mechanisms of motility in metastasizing cells. Mol Cancer Res 2010; 8(5): 629-42.[http://dx.doi.org/10.1158/1541-7786.MCR-10-0139] [PMID: 20460404][29]Cano A, Pérez-Moreno MA, Rodrigo I, et al. The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol 2000; 2(2): 76-83.[http://dx.doi.org/10.1038/35000025] [PMID: 10655586][30]Nagaraj NS, Datta PK. Targeting the transforming growth factor-beta signaling pathway in human cancer. Expert Opin Investig Drugs 2010; 19(1): 77-91.[http://dx.doi.org/10.1517/13543780903382609] [PMID: 20001556][31]Heldin CH, Landström M, Moustakas A. Mechanism of TGF-beta signaling to growth arrest, apoptosis, and epithelial-mesenchymal transition. Curr Opin Cell Biol 2009; 21(2): 166-76.[http://dx.doi.org/10.1016/j.ceb.2009.01.021] [PMID: 19237272][32]Nye MD, Almada LL, Fernandez-Barrena MG, et al. The transcription factor GLI1 interacts with SMAD proteins to modulate transforming growth factor β-induced gene expression in a p300/CREB-binding protein-associated factor (PCAF)-dependent manner. J Biol Chem 2014; 289(22): 15495-506.[http://dx.doi.org/10.1074/jbc.M113.545194] [PMID: 24739390][33]Valcourt U, Kowanetz M, Niimi H, Heldin CH, Moustakas A. TGF-beta and the Smad signaling pathway support transcriptomic reprogramming during epithelial-mesenchymal cell transition. Mol Biol Cell 2005; 16(4): 1987-2002.[http://dx.doi.org/10.1091/mbc.e04-08-0658] [PMID: 15689496][34]LoPiccolo J, Blumenthal GM, Bernstein WB, Dennis PA. Targeting the PI3K/Akt/mTOR pathway: effective combinations and clinical considerations. Drug Resist Updat 2008; 11(1-2): 32-50.[http://dx.doi.org/10.1016/j.drup.2007.11.003] [PMID: 18166498][35]Engelman JA. Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer 2009; 9(8): 550-62.[http://dx.doi.org/10.1038/nrc2664] [PMID: 19629070][36]Ghayad SE, Cohen PA. Inhibitors of the PI3K/Akt/mTOR pathway: new hope for breast cancer patients. Recent Patents Anticancer Drug Discov 2010; 5(1): 29-57.[http://dx.doi.org/10.2174/157489210789702208] [PMID: 19751211][37]Gavert N, Ben-Ze’ev A. Epithelial-mesenchymal transition and the invasive potential of tumors. Trends Mol Med 2008; 14(5): 199-209.[http://dx.doi.org/10.1016/j.molmed.2008.03.004] [PMID: 18406208][38]Pezzella F, Harris AL, Tavassoli M, Gatter KC. Blood vessels and cancer much more than just angiogenesis. Cell Death Discov 2015; 1(1): 15064.[http://dx.doi.org/10.1038/cddiscovery.2015.64] [PMID: 27551488][39]Bielenberg DR, Zetter BR. The contribution of angiogenesis to the process of metastasis. Cancer J 2015; 21(4): 267-73.[http://dx.doi.org/10.1097/PPO.0000000000000138] [PMID: 26222078][40]Benazzi C, Al-Dissi A, Chau CH, et al. Angiogenesis in spontaneous tumors and implications for comparative tumor biology. Scientific World Journal 2014; 2014: 919570.[http://dx.doi.org/10.1155/2014/919570] [PMID: 24563633][41]Schneider BP, Miller KD. Angiogenesis of breast cancer. J Clin Oncol 2005; 23(8): 1782-90.[http://dx.doi.org/10.1200/JCO.2005.12.017] [PMID: 15755986][42]Shibuya M. Vascular endothelial growth factor (VEGF) and its receptor (VEGFR) signaling in angiogenesis: a crucial target for anti- and pro-angiogenic therapies. Genes Cancer 2011; 2(12): 1097-105.[http://dx.doi.org/10.1177/1947601911423031] [PMID: 22866201][43]Zhan P, Ji YN, Yu LK. VEGF is associated with the poor survival of patients with prostate cancer: a meta-analysis. Transl Androl Urol 2013; 2(2): 99-105.[PMID: 26816732][44]Niu G, Chen X. Vascular endothelial growth factor as an anti-angiogenic target for cancer therapy. Curr Drug Targets 2010; 11(8): 1000-17.[http://dx.doi.org/10.2174/138945010791591395] [PMID: 20426765][45]Ferrara N, Alitalo K. Clinical applications of angiogenic growth factors and their inhibitors. Nat Med 1999; 5(12): 1359-64.[http://dx.doi.org/10.1038/70928] [PMID: 10581076][46]Shibuya M. Structure and function of VEGF/VEGF-receptor system involved in angiogenesis. Cell Struct Funct 2001; 26(1): 25-35.[http://dx.doi.org/10.1247/csf.26.25] [PMID: 11345501][47]Wu Y, Hooper AT, Zhong Z, et al. The vascular endothelial growth factor receptor (VEGFR-1) supports growth and survival of human breast carcinoma. Int J Cancer 2006; 119(7): 1519-29.[http://dx.doi.org/10.1002/ijc.21865] [PMID: 16671089][48]Schmidt M, Voelker H-U, Kapp M, Dietl J, Kammerer U. Expression of VEGFR-1 (Flt-1) in breast cancer is associated with VEGF expression and with node-negative tumour stage. Anticancer Res 2008; 28(3A): 1719-24.[PMID: 18630531][49]Bando H, Weich HA, Brokelmann M, et al. Association between intratumoral free and total VEGF, soluble VEGFR-1, VEGFR-2 and prognosis in breast cancer. Br J Cancer 2005; 92(3): 553-61.[http://dx.doi.org/10.1038/sj.bjc.6602374] [PMID: 15668703][50]Yao J, Wu X, Zhuang G, et al. Expression of a functional VEGFR-1 in tumor cells is a major determinant of anti-PlGF antibodies efficacy. Proc Natl Acad Sci USA 2011; 108(28): 11590-5.[http://dx.doi.org/10.1073/pnas.1109029108] [PMID: 21709213][51]Kosaka Y, Kataoka A, Yamaguchi H, et al. Vascular endothelial growth factor receptor-1 mRNA overexpression in peripheral blood as a useful prognostic marker in breast cancer. Breast Cancer Res 2012; 14(5): R140.[http://dx.doi.org/10.1186/bcr3345] [PMID: 23113927][52]Mercurio AM, Lipscomb EA, Bachelder RE. Non-angiogenic functions of VEGF in breast cancer. J Mammary Gland Biol Neoplasia 2005; 10(4): 283-90.[http://dx.doi.org/10.1007/s10911-006-9001-9] [PMID: 16924371][53]Salameh A, Galvagni F, Bardelli M, Bussolino F, Oliviero S. Direct recruitment of CRK and GRB2 to VEGFR-3 induces proliferation, migration, and survival of endothelial cells through the activation of ERK, AKT, and JNK pathways. Blood 2005; 106(10): 3423-31.[http://dx.doi.org/10.1182/blood-2005-04-1388] [PMID: 16076871][54]Liang Y, Brekken RA, Hyder SM. Vascular endothelial growth factor induces proliferation of breast cancer cells and inhibits the anti-proliferative activity of anti-hormones. Endocr Relat Cancer 2006; 13(3): 905-19.[http://dx.doi.org/10.1677/erc.1.01221] [PMID: 16954439][55]Balkwill FR, Capasso M, Hagemann T. The tumor microenvironment at a glance. J Cell Sci 2012; 125(Pt 23): 5591-6.[http://dx.doi.org/10.1242/jcs.116392] [PMID: 23420197][56]Hass R, Otte A. Mesenchymal stem cells as all-round supporters in a normal and neoplastic microenvironment. Cell Commun Signal 2012; 10(1): 26.[http://dx.doi.org/10.1186/1478-811X-10-26] [PMID: 22943670][57]Li HJ, Reinhardt F, Herschman HR, Weinberg RA. LiHJCancer-stimulated mesenchymal stem cells create a carcinoma stem cell niche via prostaglandin E2 signaling. Cancer Discov 2012; 2(9): 840-55.[http://dx.doi.org/10.1158/2159-8290.CD-12-0101] [PMID: 22763855][58]Melzer C, von der Ohe J, Lehnert H, Ungefroren H, Hass R. Cancer stem cell niche models and contribution by mesenchymal stroma/stem cells. Mol Cancer 2017; 16(1): 28.[http://dx.doi.org/10.1186/s12943-017-0595-x] [PMID: 28148265][59]Melzer C, Yang Y, Hass R. Interaction of MSC with tumor cells. Cell Commun Signal 2016; 14(1): 20.[http://dx.doi.org/10.1186/s12964-016-0143-0] [PMID: 27608835][60]Ungefroren H, Sebens S, Seidl D, Lehnert H, Hass R. Interaction of tumor cells with the microenvironment. Cell Commun Signal 2011; 9(1): 18.[http://dx.doi.org/10.1186/1478-811X-9-18] [PMID: 21914164][61]Sakakura T, Nishizuka Y, Dawe CJ. Mesenchyme-dependent morphogenesis and epithelium-specific cytodifferentiation in mouse mammary gland. Science 1976; 194(4272): 1439-41.[http://dx.doi.org/10.1126/science.827022] [PMID: 827022][62]Maller O, Martinson H, Schedin P. Extracellular matrix composition reveals complex and dynamic stromal-epithelial interactions in the mammary gland. J Mammary Gland Biol Neoplasia 2010; 15(3): 301-18.[http://dx.doi.org/10.1007/s10911-010-9189-6] [PMID: 20811805][63]Feng Y, Spezia M, Huang S, et al. Breast cancer development and progression: Risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes Dis 2018; 5(2): 77-106.[http://dx.doi.org/10.1016/j.gendis.2018.05.001] [PMID: 30258937][64]Orimo A, Gupta PB, Sgroi DC, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 2005; 121(3): 335-48.[http://dx.doi.org/10.1016/j.cell.2005.02.034] [PMID: 15882617][65]Xing F, Saidou J, Watabe K. Cancer associated fibroblasts (CAFs) in tumor microenvironment. Front Biosci 2010; 15(1): 166-79.[http://dx.doi.org/10.2741/3613] [PMID: 20036813][66]Chakravarthi BV, Nepal S, Varambally S. Genomic and Epigenomic Alterations in Cancer. Am J Pathol 2016; 186(7): 1724-35.[http://dx.doi.org/10.1016/j.ajpath.2016.02.023] [PMID: 27338107][67]Chabottaux V, Noel A. Breast cancer progression: insights into multifaceted matrix metalloproteinases. Clin Exp Metastasis 2007; 24(8): 647-56.[http://dx.doi.org/10.1007/s10585-007-9113-7] [PMID: 17968664][68]Man YG, Sang QX. The significance of focal myoepithelial cell layer disruptions in human breast tumor invasion: a paradigm shift from the “protease-centered” hypothesis. Exp Cell Res 2004; 301(2): 103-18.[http://dx.doi.org/10.1016/j.yexcr.2004.08.037] [PMID: 15530847][69]Ruffell B, DeNardo DG, Affara NI, Coussens LM. Lymphocytes in cancer development: polarization towards pro-tumor immunity. Cytokine Growth Factor Rev 2010; 21(1): 3-10.[http://dx.doi.org/10.1016/j.cytogfr.2009.11.002] [PMID: 20005150][70]Zamarron BF, Chen W. Dual roles of immune cells and their factors in cancer development and progression. Int J Biol Sci 2011; 7(5): 651-8.[http://dx.doi.org/10.7150/ijbs.7.651] [PMID: 21647333][71]Schedin P, O’Brien J, Rudolph M, Stein T, Borges V. Microenvironment of the involuting mammary gland mediates mammary cancer progression. J Mammary Gland Biol Neoplasia 2007; 12(1): 71-82.[http://dx.doi.org/10.1007/s10911-007-9039-3] [PMID: 17318269][72]Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol 2008; 9(5): 503-10.[http://dx.doi.org/10.1038/ni1582] [PMID: 18425107][73]Hacene K, Desplaces A, Brunet M, Lidereau R, Bourguignat A, Oglobine J. Competitive prognostic value of clinicopathologic and bioimmunologic factors in primary breast cancer. Cancer 1986; 57(2): 245-50. [PMID: 3484657][74]Malladi S, Macalinao DG, Jin X, et al. Metastatic Latency and Immune Evasion through Autocrine Inhibition of WNT. Cell 2016; 165(1): 45-60.[http://dx.doi.org/10.1016/j.cell.2016.02.025] [PMID: 27015306][75]Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009; 9(3): 162-74.[http://dx.doi.org/10.1038/nri2506] [PMID: 19197294][76]Ostrand-Rosenberg S, Sinha P. Myeloid-derived suppressor cells: linking inflammation and cancer. J Immunol 2009; 182(8): 4499-506.[http://dx.doi.org/10.4049/jimmunol.0802740] [PMID: 19342621][77]Safarzadeh E, Hashemzadeh S, Duijf PHG, et al. Circulating myeloid-derived suppressor cells: An independent prognostic factor in patients with breast cancer. J Cell Physiol 2019; 234(4): 3515-25.[http://dx.doi.org/10.1002/jcp.26896] [PMID: 30362521][78]Mandó P, Rizzo M, Roberti MP, et al. High neutrophil to lymphocyte ratio and decreased CD69+NK cells represent a phenotype of high risk in early-stage breast cancer patients. OncoTargets Ther 2018; 11: 2901-10.[http://dx.doi.org/10.2147/OTT.S160911] [PMID: 29844687][79]Lewis CE, Leek R, Harris A, McGee JO. Cytokine regulation of angiogenesis in breast cancer: the role of tumor-associated macrophages. J Leukoc Biol 1995; 57(5): 747-51.[http://dx.doi.org/10.1002/jlb.57.5.747] [PMID: 7539028][80]Hagemann T, Wilson J, Kulbe H, et al. Macrophages induce invasiveness of epithelial cancer cells via NF-kappa B and JNK. J Immunol 2005; 175(2): 1197-205.[http://dx.doi.org/10.4049/jimmunol.175.2.1197] [PMID: 16002723][81]