Animal Models in Experimental Medicine -  - E-Book

Animal Models in Experimental Medicine E-Book

0,0
54,93 €

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

This reference presents information about models utilized in experimental medicine and pharmaceutical research and development for several human diseases. Written by experts in immunology, cancer biology and pharmacology, the book provides readers with handy notes and updated data on animal models that are critical to research planning and lab execution.

The main feature of the book is a set of 12 structured chapters that focus on a specific disease such as cancer, infectious diseases, autism, autoimmune diseases, Alzheimer’s disease and anemia. The contributors have gathered information on a wide range of genetic and physiological animal models that are employed in research with comparative charts that highlight their main differences. The book also includes chapters for special topics like food allergies and dentistry. Additional features of the book are an explanation of disease mechanisms that give an easy understanding, notes for idiopathic models and specific clinical conditions, and a list of references for advanced readers.

Animal Models in Experimental Medicine is essential reading for scholars, graduate students and senior researchers in life sciences and clinical medicine. It also serves as a resource for professionals involved in bench-to-bedside pharmaceutical projects.

Readership
Scholars, graduate students and senior researchers in life sciences and clinical medicine; professionals involved in bench-to-bedside pharmaceutical projects.

Das E-Book können Sie in Legimi-Apps oder einer beliebigen App lesen, die das folgende Format unterstützen:

EPUB
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:
PREFACE
DEDICATION
List of Contributors
Animal Models for Cancer
Abstract
Introduction
Mechanisms of cancer disease
Cell Cycle
Characteristics of Developing Tumors
Escaping or Evading Growth Suppressors
Ability to Enable Replicative Immortality
Capacity to Sustain Proliferative Signalling
Genome Instability and Mutations
Capability for Resisting Apoptosis
Tumour-promoting Inflammation
Triggering Invasion and Metastasis
Capability for Inducing/accessing the Vasculature
Avoiding Immune Destruction System
Cellular Metabolism Reprogramming
Animal models in cancer research
Mice
Chemically Induced Mice
Genetically Engineered Mice
Patient-derived Tumor Xenograft
Zebrafish
Drosophila
Non-human Primates
Conclusion
References
Animal Modeling of Infectious Diseases
Abstract
Introduction
Schistosomiasis Model
Ascaris Lumbericoides Model
Fasciola Hepatica Model
Hepatitis C Virus Model
Listeria Monocytogenes Model
Entamoeba Histolytica Model
Cytomegalovirus Animal Model
NON-PRIMATE SPECIES
Non-Human Primates
Conclusion
References
Autoimmune Diseases in Animals
Abstract
TYPE-1 DIABETES MELLITUS, NON-OBESE DIABETIC (T1DM) IN ANIMAL MODELS
Introduction
Epidemiology of Type-1 Diabetes Mellitus
Risk Factors for Type-1 Diabetes Mellitus
Animal Models of Type-1 Diabetes Mellitus
Non-obese Diabetic Mouse (NOD)
Genetically Modified Mice Models
Biobreeding (BB) Rat
Komeda Diabetes Prone (KDP) Rat
Conclusion
TYPE 2 DIABETES MELLITUS (T2DM) IN ANIMAL MODELS
Introduction
Disease Models Listed by Animals in the Past
CABC in Large Animals
Differences from Human Models
AUTOIMMUNE THYROID DISEASES IN ANIMAL MODELS
Introduction
Epidemiology of Autoimmune Thyroid Diseases
Risk Factors for Autoimmune Thyroid Diseases
Possible Mechanism of Disease Development
Animal Models of Human AITD
The Spontaneous AITD Animal Model
Induced AITD Model
Conclusion
ARTHRITIS IN ANIMAL MODELS
Introduction
The Animal Models Used
Disease Mechanism
Rat Adjuvant Arthritis
The Cytokine Responses During the Course of Arthritis
The Features of each Model
DIFFERENCES FROM HUMAN MODELS
In Vivo Models for RA
Conclusion
REFRENCES
Animal Models of Anemia
Abstract
Introduction
General Overview
Animal Models of Anemia of Inflammation
The Mouse Models of Anemia of Inflammation
Transgenic Models Anemia of Inflammation
The Rats’ Models of Anemia of Inflammation
Fish Models of Anemia of Inflammation
Animal Models of Iron Deficiency Anemia During Pregnancy
Animal Models of Iron Overload Hemochromatosis (HH)
Animal Models of Menkes Syndrome
Animal Models of Wilson Disease
Conclusion
References
Animal Models of Alzheimer's Disease
Abstract
Introduction
Mice
PDAPP
Tg2576
APP23
TgCRND8
APP/PS1
3xTg-AD
5xFAD
APPNL-G-F Knock-in Mice
APOE4-TR
TREM2 KO/APPPS1
PS19
ALDH2 KO
Injury-based and Trauma-based Models
Other KO Mice
Chemically Induced AD Models
Mouse Model Drawbacks
RATS
Familial Swedish and Indiana
PSAPP (Tg478/Tg1116/Tg11587)
AD tau
Chemically-induced
LARGE ANIMALS
Non-human Primates
CONCLUSION
References
Animal Models of Asthma
Abstract
Introduction
Asthma Phenotypes/endotypes
Animal Models and Asthma: Value, Criteria of Selection, and Limitations
Murine Models: Investigation of Asthma Pathogenesis and Airway Remodeling
Murine Models: Investigation of the Genetic Basis of Asthma
Murine Models: Role in Drug Discovery for Asthma
Murine Models in Asthma: Limitations and Validity of Data Extrapolation
Non-murine Models for Asthma
Conclusion
References
Atherosclerosis in Animals
Abstract
Introduction
DISEASE MECHANISM
Disease Models Listed by Animals
FEATURES OF EACH MODEL
Rabbit Model
Rat Model
Pig and Non-human Model
Other Animal Models of Atherosclerosis
Difference from Human Models
Conclusion
References
Animal Models of Food Allergy
Abstract
Introduction
Review of food hypersensitivity Pathogenesis
Animal Models of Food Allergy
Murine Model
Mouse Model with Adjuvant
Food Protein Allergen
Adjuvant Type
Route of Administration
Dose of Food Protein and Adjuvant Doses
Number of Doses During and Length of the Sensitization Phase and Challenge Dose
Mouse Model Without Adjuvant
Genetically Modified Mice
Transgenic/Humanized Mouse Model
RAT Food Allergy Model
Guinea Pig Model
Canine Model
Swine Model
Sheep Model
Conclusion
References
Animal Models in Dentistry
Abstract
Introduction
DISEASE MECHANISM
Periodontal Disease
Dental Caries
Oral Cancer
DISEASE MODEL LISTED BY ANIMALS
ANIMAL MODELS
Features of Each Animal Models used in Periodontology
Dogs
Cats
Rats
Hamsters
Minks
Miniature Pigs
Mice
Baker Mouse Model
Non-Human Primates
Other Animals
Rabbit
Horses
Features of Each Model Used in Oral Cancer
Rats Model
Mouse Model
Other Models
Hamster Model
Primates Model
Features of Each Model used in Dental Caries
Rodent Model
Rat Model
Hamster Model
Pig Model
Dog Model
Other Models
Non-Human Primates
Difference from Human Model
Conclusion
References
Animal Models for the Study of Autism
Abstract
Introduction
ANIMAL MODELS OF AUTISM
Genetic Models for Autism
Animal Models for Genes of Human Syndromic Disorders Predisposing to Autism
Fragile Mental Retardation 1 (FMR1) Gene Model
Methyl CpG Binding Protein 2 (MECP2) Gene Model
SH3 and Multiple Ankyrin Repeat Domains Protein 3 (SHANK3) Gene Model
Tuberous Sclerosis Complex 1/2 (TSC1/2) Gene Model
15q11-q13 Gene Locus Mutation Model
16p11.2 Gene Locus Mutation Model
Animal Models for Non-syndromic Genes Associated with Autism
Neuroligin (NLGN) Genes Model
Neurexin (NRXN) Genes Model
Idiopathic Model of Autism
Environment Exposure Models for Autism
Toxin-induced Animal Models of Autism
Methylmercury (MeHg) Induced Model
Polychlorinated Biphenyls (PCBs) Induced Model
Bisphenol Propane (BPA) Induced Model
Drug-induced Animal Models of Autism
Sodium Valproate (VPA) Induced Model
Propionic Acid-induced Model (PPA)
Other Environment-induced Models of Autism
Prenatal Stress-induced Models
Gut Microbiota (GM) Models
Maternal Autoantibodies-related Autism Model
Maternal Immune Activation (MIA)
Characterization of Animal Models of Autism
Conclusion
References
Animal Models for the Study of Osteoporosis
Abstract
Introduction
Osteoporosis
Animal Models of Osteoporosis
Specific Animal Models
Primary Osteoporosis Model: Ovariectomy induced Animal Model for Postmenopausal Osteoporosis
Rodent Model for Postmenopausal Osteoporosis
Rabbit Model for Postmenopausal Osteoporosis
Sheep Model for Postmenopausal Osteoporosis
Other Large Animal Models for Postmenopausal Osteoporosis
Primary Osteoporosis Model: Senile Osteoporosis
Secondary Osteoporosis Model
Glucocorticoid-Induced Osteoporosis Model
Small Animal Models for Glucocorticoid-induced Osteoporosis
Large Animal Models for Glucocorticoid Induced Osteoporosis
Retinoic Acid-induced Osteoporosis Model
Alcoholic Osteoporosis Model
Disuse Osteoporosis Model
Methods for analysis/screening of animal Models of osteoporosis
Dual Energy X-ray Absorptiometry (DXA)
Peripheral Quantitative Computed Tomography (Pqct) and Microcomputed Tomography (µCT)
Bone Histomorphometry
Serum Markers of Bone Turnover
Conclusion
References
Experimental Models in Autoimmune Uveitis
Abstract
Introduction
Autoimmune Uveitis (AU)
Etiopathogenesis of Autoimmune Uveitis
ANIMAL MODEL FOR EXPERIMENTAL AUTOIMMUNE UVEITIS
Retinal S-Antigen (S-Ag)
Interphotoreceptor Retinoid-Binding Protein (IRBP)
Recoverin
Rhodopsin
Phosduscin
Retinal Heat Shock Protein 60 (HSP60)
EXPERIMENTAL AUTOIMMUNE UVEITIS [EAU]
Induced Animal Models of Uveitis
“Classical” EAU Induced in B10.RIII, B10. A and C57BL/6 mice
EAU Induced by Retinal Antigens in Lewis Rat Model
EAU Induced by Antigen-pulsed Syngeneic Dendritic Cells in Mice
Pain and Distress Assessment
Ethical Oversight
Alternative Adjuvants
Dosage Optimization/Standardization
Refinement of Techniques
Spontaneous Uveitis Models
B10.RIII Mouse Model
Autoimmune Regulator (AIRE) Knockout Mice
Transgenic Models
Humanized Animal Models of Uveitis
THE EXPERIMENTAL AUTOIMMUNE UVEITIS (EAU) GRADING SYSTEM
Conclusion
References
Animal Models in Experimental Medicine
Edited by
Faris Q. Alenzi
College of Applied Medical Sciences
Prince Sattam bin Abdulaziz University (PSAU)
Alkharj, Saudi Arabia

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 book/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 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 Singapore. Each party agrees that the courts of the state of Singapore 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 Pte. Ltd. 80 Robinson Road #02-00 Singapore 068898 Singapore Email: [email protected]

PREFACE

For both experimental and clinical trials, animal models have been used extensively for many years. The research is currently focused on many approaches that hold great promise by offering radical new successful treatments for many human diseases by using these animal models. The current book provides a summary of the current knowledge and the use of these models as well as their pre-clinical implications and related therapeutic options. Furthermore, the recently discovered trials of these animals in cancer, autoimmunity, infectious diseases, allergy, dental and other diseases have encouraged us to write this book in depth for developing new pharmacological and immunological treatments and for a better understanding of many chemo-resistant drugs.

Faris Q. Alenzi College of Applied Medical Sciences Prince Sattam bin Abdulaziz University (PSAU) Alkharj, Saudi Arabia

DEDICATION

I dedicate this book to who inspired me

To,

Adnan Albar

John Goldman

Myrtle Gordon

Abdulla Alnaser

George Eyambe

Richard Wyse

Anthony Warrens

Robert Lechler

Mohamed Salem

Abdulla Alothman

List of Contributors

Adel M. AlqarniDepartment of Clinical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Alkharj-Saudi ArabiaAisha Al AnaziConsultant Pediatrics Clinical Endocrinology and Diabetes, Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi ArabiaAbdulaziz Al AniziKAMC, Riyadh, Saudi ArabiaAmmar Y. JastaniahCollege of Pharmacy, Al-Faisal University, Riyadh-11533, Saudi ArabiaAhmed M. Al-HakamiDepartment of Microbiology and Clinical Parasitology, College of Medicine, King Khalid University, Abha-62529, Saudi ArabiaAnandhalakshmi SubramanianDepartment of Microbiology and Clinical Parasitology, College of Medicine, King Khalid University, Abha-62529, Saudi ArabiaDeema I. FallatahDepartment of Clinical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Alkharj, 11942, Saudi ArabiaFahad N. AlonaziPublic Health Laboratories, Public Health Authority, Riyadh-11533, Saudi ArabiaFaihan Al AniziKAMC, Riyadh, Saudi ArabiaFahad E. AlbisiKAMC, Riyadh, Saudi ArabiaGhfren AlorainiDepartment of Clinical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University (PSAU), Alkharj, Saudi ArabiaHelal G. AlanaziDepartment of Clinical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Alkharj, Saudi ArabiaHuda Abdullah AlmutairiSpecialist in Prosthodontics, Dental Department, King Saud Hospital, Qassim Province, Unaizah, Saudi ArabiaJana Ali AlnajimSpecialist in Paediatric Dentistry, Dental Department, King Saud Hospital, Qassim Province, Unaizah, Saudi ArabiaKhalid Jamal AlanaziPedodontist, Dental Center, Alyamamah Hospital, Riyadh Province, Riyadh, Saudi ArabiaMohammed A. AfifiDepartment of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi ArabiaMohammed W. Al-RabiaDepartment of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi ArabiaMona AlanaziCollege of Nursing, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia King Abdullah International Medical Research Center, Riyadh, Saudi ArabiaNoufa Al OnaziConsultant Pediatrics Allergy and Clinical Immunology, Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi ArabiaRakan J. AlanaziDepartment of Pharmacy Practice, College of Pharmacy, Al-Faisal University, Riyadh, Saudi ArabiaSuad A. AlghamdiDepartment of Clinical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Alkharj-Saudi ArabiaSaba AbdiDepartment of Biochemistry, College of Science, King Saud University, Riyadh-11495, Saudi ArabiaTalal Adnan BashaPediatric Allergy and Immunology Consultant, Department of Pediatric, Faculty of Medicine, University of Jeddah, Jeddah, Saudi ArabiaYousef HawsawiDepartment of Epidemiology and Statistics, Research Center, King Faisal Specialist Hospital and Research Center, Jeddah-21499, Kingdom of Saudi Arabia Department of Biochemistry & Molecular Medicine, College of Medicine, Al-Faisal University, Riyadh-11533, Saudi Arabia

Animal Models for Cancer

Fahad N. Alonazi1,*,Yousef M. Hawsawi2,3,Helal G. Alanazi4,Adel M. Alqarni4,Suad A. Alghamdi4,Rakan J. Alanazi5
1 Public Health Laboratories, Public Health Authority, Riyadh-11533, Saudi Arabia
2 Research Centre at King Faisal Specialist Hospital and Research Center, Jeddah-21499, Saudi Arabia
3 Department of Biochemistry & Molecular Medicine, College of Medicine, Al-Faisal University, Riyadh-11533, Saudi Arabia
4 Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Alkharj-Saudi Arabia
5 Department of Pharmacy Practices, College of Pharmacy, Alfaisal University, Riyadh-11533, Saudi Arabia

Abstract

Cancer is a complex multifactorial disease that affects many people worldwide. Animal models play an important role in deciphering cancer biology and developing new therapies. The animal models widely used in cancer research include tumor xenografts, genetically engineered mice, chemically induced models, and spontaneous tumor models. These models provide a controlled environment to study cancer progression, the interaction of cancer and the immune system, and the effectiveness of new therapies. Although animal models have several advantages, it is important to identify their limitations and use them in conjunction with other preclinical models, such as in-vitro cell culture and patient-derived xenografts, to ensure that results are transferable to humans. In this chapter, we discuss the importance of animal models in cancer research, the different types of animal models, and their advantages and disadvantages. We also provide some examples of animal models used in cancer research. Collectively, animal models have been invaluable in advancing our understanding of cancer and will continue to be important tools in the development of new therapies.

Keywords: Animal models, Advantages, Cancer, Combination therapies, Chemically induced models, Genetically engineered mice, Immune system, KRAS, Lung cancer, Limitations, Off-target effects, Pancreatic cancer, Spontaneous tumor models, Tumor xenografts, Transgenic mice.
*Corresponding author Fahad N. Alonazi: Public Health Laboratories, Public Health Authority, Riyadh, 11533, Saudi Arabia; E-mail: [email protected]

Introduction

Cancer is a leading killer worldwide, and despite significant advances in diagnosis and treatment, it remains a poorly understood and treated disease. One of the reasons for this challenge is the complexity of the disease, involving multiple genetic and environmental factors [1]. Animal models play a crucial role in advancing our understanding of cancer biology and the advancement of new therapies. They provide a controlled environment to study cancer progression, its interaction with the immune system and its response to various treatments. Animal models are also useful for identifying new therapeutic targets and understanding mechanisms of drug resistance.

In recent years, advances in genetic engineering have enabled the creation of animal models that better mimic human cancers, such as B. mice with mutations in the KRAS oncogene. These models provide valuable insights into mechanisms of carcinogenesis and have been used to test new therapies targeting specific molecular pathways.

Despite advances in cancer research using animal models, there are still challenges and limitations that need to be addressed. Off-target effects, differences in the tumor microenvironment, and lack of representation of human diversity are just some of the challenges researchers face. Future research should focus on improving animal models to better mimic human disease, identifying biomarkers of treatment response, and developing combination therapies targeting multiple oncogenic pathways.

Mechanisms of cancer disease

The adult human body is composed of approximately 37 trillion cells, expressed in various sizes, types and shapes, which together define the structure and function of the entire body. Although most cells are regularly damaged by wear and tear, their numbers remain strictly constant. This is due to the highly precise regulation of cell division and differentiation processes (i.e., cellular homeostasis between cell death and reproduction). Disruption of this balance and failure of apoptosis can lead to aggressive and uncontrolled cell division, ultimately leading to the development of cancer.

Cancer refers to a group of diseases characterized by the uncontrolled division of cells. Cancer can strike anyone, regardless of sex, age, and social status. It can start in any part or organ of the body and can be classified according to the tissue of origin. The transformation of normal cells into cancer cells involves alterations in the action of different genes and proteins in the cells. Under the influence of certain conditions, cancer cells can spread from the tissue of origin to nearby tissues and other non-adjacent tissues (metastasis), interfering with their normal functions.

Irrespective of technological advances, breast cancer originating in breast tissue remains one of the leading causes of cancer-related death in women worldwide [2]. Cancer genetics have revolutionized our understanding of breast cancer. The discovery of complementary DNA (cDNA) array analysis in the past century was a remarkable breakthrough in breast cancer research to evaluate the effect of a precise tumor signature on prognosis [3]. This innovation opens a new window into the molecular classification of breast cancer. Currently, the progression of breast cancer is highly dependent on altering the expression of multiple genes. To date, more than 8,000 genes have been found to be differentially expressed in different breast cancer subtypes [4], hence the concept of genotype-phenotype correlation.

However, immunohistochemical studies distinguished four distinct molecular subtypes of breast cancer, including normal breast cancer, HER-2-positive, basal-like, and luminal-like. Luminal A and Luminal B are additional classifications of the major subtypes of Luminal [5]. The most common subtype of breast cancer is Luminal A, followed by Luminal B. Her-2 is expressed in approximately 40% of breast cancers, with even expression of hormone receptor-positive and -negative subtypes. A small proportion (10%) of breast cancer tumors are basal-like tumors, also known as triple-negative (TN) breast cancers due to their molecular makeup. With a brief background on the classification of breast cancer, we can now turn to the mechanisms of the disease.

Breast cancer is a heterogeneous disease caused by multiple factors through multiple mechanisms. This fatal disease develops over many years and through multiple stages. The multistep progression of carcinogenesis is shaped by complementary mechanisms known as cancer hallmarks. The concept of a “cancer hallmark” refers to a set of functional capabilities that human cancer cells acquire as they progress from a normal growth phase to a neoplastic growth phase. Especially skills that are crucial for the development of malignancies. In early 2000, Hanahan and Weinberg enumerated their hypothesized universal features that link all the different cancer cells at the cellular phenotype level [6, 7]. Initially, they predicted the contribution of different hallmark abilities and later expanded the different hallmarks to eight [7].

The hallmarks include avoiding growth inhibitors, sustaining proliferative signals, promoting angiogenesis, achieving replicative immortality, and inducing invasion and metastasis [6]. Kim and colleagues also reported that genome stability and cancer-associated inflammation are additional features associated with tumorigenesis. It is worth showing some important aspects of the cell cycle to understand the processes of the various hallmarks.

Cell Cycle

Understanding the cell cycle, which regulates DNA replication and cell division, is critical to understanding the molecular pathways of cancer [8]. Five dynamic phases are critical during the cell cycle. The first phase, the interphase, consists of three separate consecutive phases. The first phase is the G1 “monitoring” phase, followed by the S phase, in which DNA is synthesized, and finally, the third phase, G2, when cells continue to develop and prepare for mitosis. Thereafter, cells enter a critical phase called the mitosis (M) phase [9]. There are four phases of mitosis: telophase, anaphase, metaphase, and prophase. Cells can re-enter the cell cycle and undergo DNA replication and mitosis during quiescence (G0), a biochemically distinct state.

Changes in the activity of certain cyclin-dependent kinases (CDKs) drive transitions between these phases, with Cdk1/Cdk2 and Cdk2/Cdk4/Cdk6 directing the transition from G2 to mitosis and G1 to S phase, respectively [9]. With this introduction, we can now understand the mechanisms of carcinogenesis and the hallmarks of cancer.

Characteristics of Developing Tumors

Cancer hallmarks currently include the following hallmarks: escape or evasion of growth inhibitors; ability to immortalize replication, ability to sustain proliferative signals, promotion of tumor inflammation, ability to resist cell death programs, induction of invasion and metastasis, genomic instability and mutation, ability to induce/reach blood vessels, immune system evasion, cellular metabolic reprogramming, cellular metabolic dysregulation [10]. These hallmarks play a crucial role in the development of cancer [11]. So, we will discuss each hallmark in detail in the following sections.

Escaping or Evading Growth Suppressors

Cancer cells can bypass growth inhibitory pathways. Returning to the cell cycle, it is important to emphasize that these components determine how cells move through the G1 phase of their developmental cycle. These processes are known as anti-growth signals and are involved in the cell cycle of normal cells. However, to survive, cancer cells resist antiproliferative signals. To understand how this process works, we need to understand an important protein called the retinoblastoma protein (pRb). pRb and its two cousins​​, p107 and p130, are molecular pathways for many if not all, antiproliferative signals. pRb inhibits proliferation when in a hypophosphorylated state by secreting and altering the function of the E2 factor (E2F) transcription factor that regulates expression of the gene pool necessary for progression from G1 to S phase [12].

When the pRb pathway is disrupted, E2F is released, which promotes cell proliferation and converts cells to growth factor-resistant cells, thereby preventing cell cycle entry into the G1 phase. Another mechanism that inhibits G1 phase progression is through TGF [13]. The TGF inhibits progression by preventing the phosphorylation that renders pRb inactive [14]. One approach to prevent differentiation directly uses the c-myc oncogene, which encodes a transcription factor. The hallmarks of cancer complement each other, making cancer cells immortal as well.

Ability to Enable Replicative Immortality

Cancer cells can convert into immortal cells. Provocatively, it appears that most tumor cell types that are grown in culture are immortalized, indicating that infinite replicative ability is a characteristic that was acquired in vivo during tumor progression [15]. Research indicated that the telomeres at the ends of chromosomes, which are made up of thousands of repeats of a brief 6 bp sequence fragment, are the counting mechanism for cell generations. However, the loss of 50–100 bp of telomeric DNA from the ends of each chromosome during each cell cycle is scientifically used to calculate the number of replicative generations. However, almost all types of cancer cells exhibit telomere maintenance; 85%–90% of them achieve this by increasing the expression of the telomerase enzyme, which adds hexanucleotide repeats to the ends of telomeric DNA [16, 17]. On the other hand, the remaining cells have developed a method of activating a mechanism known as ALT, which appears to maintain telomeres through recombination-based interchromosomal exchanges of sequence information [16]. However, one of the cancer characteristics is the ability to sustain proliferative signaling.

Capacity to Sustain Proliferative Signalling

Usually, cancer cells can sustain proliferative signaling. During tumor pathogenesis, the cell surface receptors that transmit growth-stimulating signals into the cell interior become targets of dysregulation. Many malignancies have an overexpression of growth factor (GF) receptors [18, 19], which frequently have tyrosine kinase activity in their cytoplasmic domains. The overexpression of the receptor may make cancer cells more sensitive to GF levels in the environment than they would usually be [20]. The ER+ breast cancer can be treated through hormonal therapies [21, 22]. Additionally, cancer cells can alter the types of extracellular matrix receptors (integrins) that they express, favoring those that provide signals for proliferative proliferation [23]. Although breast cancer development highly depends on genetic factors, genome instability and mutations are crucial.

Genome Instability and Mutations

Many studies have suggested that the malfunction of particular parts of the genomic “caretaker” systems is the cause of this mutability [24, 25]. Several genes have been associated with genome instability, including BLM and BRCA1/2 [26, 27]. However, the p53 tumor suppressor protein is the most noticeable component of these systems. When DNA is damaged, it either causes cell cycle arrest to allow for DNA repair or, if the damage is too great, the apoptosis process will be activated. It is now evident that most, if not all, cases of human malignancies lack the functionality of the p53 DNA damage signaling system [28]. It is interesting to note that current research reveals that apoptosis may also be a source of genomic instability due to the possibility of nearby cells incorporating DNA from apoptotic cell bodies after phagocytosis. Nevertheless, cancer cells can resist the cell death program. More recently, compelling evidence emerged for the concept of non-mutational epigenetic reprogramming as a cancer phenotype hallmark [29]. For instance, hypoxia decreases the activity of the TET demethylases, causing significant methylome alterations, including hypermethylation [30].

Capability for Resisting Apoptosis

Cells undergo apoptosis as part of a normal regulatory process, and often, in tumor cells these mechanisms are subverted [21]. For example, the tumor suppressor protein phosphate and tensin homolog on chromosome 10 (PTEN) is an important regulator of apoptosis, and often, within breast cancer, mutations in this protein result in the inhibition of apoptotic processes and uncontrolled cell division. Many of the signals that cause apoptosis focus on the mitochondria, which release cytochrome C, a powerful apoptosis accelerator, in response to proapoptotic signals [31]. Members of the Bcl-2 protein family, which include proteins with proapoptotic (Bax, Bak, Bid, and Bim) or antiapoptotic (Bcl-2, Bcl-XL, and Bcl-W) activity, control mitochondrial death signals [32]. When p53 detects DNA damage, it increases the production of proapoptotic Bax, which in turn prompts mitochondria to release cytochrome C [33]. Additionally, death receptors such as FAS or the cytochrome C produced from mitochondria respectively, activate two “gatekeeper” caspases, 8 and 9, respectively.

Cancer cells can develop resistance to apoptosis using a variety of techniques. Undoubtedly, the p53 tumor suppressor gene is involved in the most frequent loss of a proapoptotic regulator through mutation. More than 50% of human malignancies have this functional inactivation of the p53 protein, which removes a crucial part of the DNA damage sensor that can activate the apoptotic effector cascade [34]. In a significant portion of human malignancies, the PI3 kinase-AKT/PKB pathway, which sends antiapoptotic survival signals, is probably implicated in preventing apoptosis. Ras-derived intracellular signals [35], extracellular hormones like IGF-1/2 or IL-3 [12], or the deletion of the pTEN tumor suppressor, a phospholipid phosphatase that typically dampens the AKT survival signal, can all activate this survival signaling circuit. Cancer cells can promote inflammation. So, how did this process happen?

Tumour-promoting Inflammation

In contrast to apoptosis and autophagy, necrotic cell death sends proinflammatory signals into the surrounding tissue milieu [36]. Necrotic cells can, therefore, attract immune system inflammatory cells [37]. However, considering that immune inflammatory cells can promote angiogenesis, cancer cell proliferation, and invasiveness, there is evidence that these cells may actively promote tumor growth in the context of neoplasia. Furthermore, necrotic cells can emit bioactive regulatory factors such as IL-1 that can directly drive nearby viable cells to grow, thereby accelerating the evolution of neoplastic disease [38].

Triggering Invasion and Metastasis

The invasion-metastasis cascade, which refers to the discrete processes that make up the multistep process of invasion and metastasis, has been extensively studied [39]. The change that was best understood involves cancer cells' loss of E-cadherin, a crucial cell-to-cell adhesion protein [40]. The consistent downregulation of E-cadherin and mutational inactivation of this characteristic capability in human carcinomas offered compelling evidence for this concept. This illustration shows a series of cell-biologic changes starting with local invasion and followed by intravasation by cancer cells into nearby blood and lymphatic vessels. Afterward, cancer cells transit through the lymphatic and hematogenous systems and escape from the lumina of vessels into the parenchyma of distant tissues (extravasation). Cancer cells end up with the formation of small nodules (micrometastases) and the growth of micrometastatic lesions [41]. This process moves to the next hallmark, which is inducing or accessing the vasculature.

Capability for Inducing/accessing the Vasculature

Vascular endothelial growth factor (VEGF) and fibroblast growth factors (FGF1/2) are the main examples of signals that start angiogenesis. An “angiogenic flip” from vascular quiescence appears to be the discrete phase during tumor development that confers the capacity to trigger and maintain angiogenesis. By shifting the ratio of angiogenesis inducers to opposing inhibitors, tumors appear to turn on the angiogenic switch [42]. Comparing tumors with their equivalents in normal tissue, many cancers show elevated expression of VEGF and/or FGFs. Others have downregulated the expression of endogenous inhibitors such as thrombospondin-1 or -interferon. Additionally, in some cancers, both changes may take place and are connected [43].

Avoiding Immune Destruction System

Cancer cells develop strategies to avoid being detected and eliminated by the host's immune system [7]. Cells can accomplish this, for instance, by manipulating immunological checkpoint regulation and the innate immune response using a stimulator of interferon genes (STING) [44]. It is now clear that cancers manipulate their microenvironments, notably by altering certain immune checkpoint pathways, to elude immune monitoring and attack. T cells, sometimes referred to as tumor-infiltrating lymphocytes (TIL) in malignancies, are abundantly present at the tumor site and affect overall survival. The cell surface ligands on the tumor cell cause PD-1 on the T-cell to become active. The host immune system may be bypassed by malignancies due to the upregulation of PD-L1. TIM-3 suppresses antitumor immunity by promoting T-cell fatigue [45].

Cellular Metabolism Reprogramming

Cancer cells can also reprogram cellular metabolism and deregulate the system [35]. Increased glycolysis, increased glutaminolytic flux, upregulation of amino acid and lipid metabolism, increased mitochondrial biogenesis, induction of the pentose phosphate pathway, and macromolecule biosynthesis are some of the most notable changes in tumor cell bioenergetics [46]. The Warburg effect is a phenomenon whereby cancer cells use glycolysis even under normoxic conditions [47]. Cancer cells upregulate glucose transporters, including Glut1, Glut2, Glut3, and Glut4, to uptake more glucose. Furthermore, most glycolytic enzymes' expression is noticeably increased in cancer cells [48]. Additionally, glutaminolysis is a common source of energy and metabolism for many cancers [49]. The pentose phosphate pathway (PPP) has significant effects on several cancer-related phenomena, such as metastasis, treatment resistance, invasion, and proliferation [50]. The augmentation of mitochondrial biogenesis is yet another important modification in cancer metabolism [49]. Another striking aspect of cancer metabolism is an increase in lipid metabolism [51].

Animal models in cancer research

The primary objective of incorporating animal experimentation into cancer research is to gain an in-depth understanding of the pathophysiology of numerous types of cancer, ranging from genomics and proteomics to metabolomics levels. Animal models provide controlled environments to study cancer progression, its interaction with the immune system, and its response to different treatments. They have also been instrumental in identifying new targets for therapy and understanding the mechanisms of drug resistance. For example, preclinical studies using mouse models have led to the development of numerous successful cancer therapies, comprising trastuzumab for breast cancer, imatinib (Gleevec) for chronic myelogenous leukemia, and vemurafenib (Zelboraf) for melanoma.

In many studies, targeting of potential therapeutic relevance in cancer cells was initially validated in genetically engineered mouse (GEM) tumor models. This can be accomplished by knocking out applicant genes in the germline (provided they didn’t adversely impair viability) or by utilizing conditionally floxed alleles that were ablated dependently with the growth of K-Ras oncogene activation [52] [53]. Most of these investigations have centered on proteins and enzymes that are theorized to function as downstream effector molecules of K-Ras, like those in the mitogen-activated protein kinase (MAPK) pathway [54].

Different types of animals can be employed as model organisms for these experiments. The most used animal models in cancer research will be described below:

Mice

Mice are most frequently used in cancer research, where they are used to study the genetics of cancer development, tumor growth, and response to treatment. The development of transgenic and knockout mice has enabled the manipulation of specific genes involved in cancer, allowing researchers to identify potential drug targets and evaluate new treatments [55]. For example, a mouse model of breast cancer has been used to study the effect of the Pten gene on tumor growth and metastasis. In this study, researchers found that loss of Pten leads to increased tumor growth and metastasis, suggesting that Pten may be a potential therapeutic target [56]. However, using mice as disease models has some limitations. Mice are small, which limits the types of experiments that can be performed. Additionally, mice have a shorter lifespan than humans, making it difficult to study cancers that slowly develop over time [57].

Chemically Induced Mice

Chemically induced mice models are essential in the elucidation of the fundamental signaling mechanisms, genetic and environmental factors associated with human carcinogenesis, and the screening of potential chemo-preventive agents and drugs [58]. Similar to human tumors caused by environmental factors, the tumors induced in mice through chemicals have a considerable burden of mutations. This burden leads to a diversity of genetic and epigenetic changes, which in turn affect the prognosis of individual cases, the efficacy of treatments, and the emergence of drug resistance [59].

The induction of colon and rectal tumors in mice through subcutaneous or intraperitoneal injections of 1,2-Dimethylhydrazine (DMH) has been proven to be an efficient method of carcinogenesis. The mice model of experimental colon carcinogenesis, known as the DMH model, has been thoroughly established and is widely used. It shares many morphological and molecular similarities with sporadic colorectal cancer (CC) in humans [60]. In a skin cancer study, researchers analyzed the ability of Withaferin A (WA) to prevent the spread of cancerous cells in a chemically induced mouse model. The findings of Li et al. in 2016 revealed that WA effectively hindered skin carcinogenesis by restraining cell proliferation, as opposed to promoting cell death [61]. In general, carcinomas that are caused by chemical exposure tend to have a lack of invasiveness, both locally and distantly, with metastases being a rare occurrence. Additionally, there is often a significant delay between the initial application of the chemical and the development of the tumor in many cases [62].

Genetically Engineered Mice

The preferred method of analyzing the intrinsic and extrinsic cellular processes involved in cancer initiation, progression, and metastasis is through the use of genetically engineered mouse models (GEMMs) of de novo tumorigenesis in vivo systems [63]. The benefits of GEMMs stem from the fact that the mouse genome shares a 99% similarity with the human genome. Additionally, the small size of these animals and the availability of an extensive array of molecular tools renders them ideal candidates for conducting large-scale research studies, which can ultimately prove to be cost-effective [64]. Chemoprevention research has utilized GEMMs to a great extent, leading to a substantial contribution in comprehending the onset of tumor formation. Specifically, GEMMs have been instrumental in shedding light on early events that lead to tumor initiation [62].

Although GEMMs have their benefits, they may not consistently replicate the heterogeneity of tumors that are commonly found in humans [63]. Nonetheless, GEMMs have demonstrated potential in forecasting the response of the clinical setting when it comes to drug development. Despite this, pharmaceutical companies have been unwilling to use GEMMs due to the challenges that come with overseeing preclinical experiments. These trials often necessitate the growth of tumors for several months before a therapeutic intervention can be introduced [65].

Patient-derived Tumor Xenograft

The high failure rates in clinical trials emphasize the necessity for improved preclinical efficacy models to better predict clinical outcomes. Human preclinical models have been developed to address this issue, including the patient-derived tumor xenograft (PDX) model, which has improved our understanding of cancer growth pathways and served as an effective tool for innovative cancer therapies [66]. PDX models are generated by directly implanting or injecting human cancerous tissues into an immune-deficient mouse, providing a high degree of predictability and rapidity of tumor formation compared to genetically engineered models. Also, based on the cell lines and number of injected cancerous cells, xenografts might take less time to develop tumors [67].

Histological evaluation of tumors developed in mice compared to the corresponding original tumor showed significant conservation of morphologic properties between the two tumors. Xenograft models are also useful for testing cancer therapies and maximizing the outcomes of these models random transgene integration can also result in unexpected phenotypes, and it is difficult to control the level and pattern of gene expression in GEMMs chemopreventive therapies, leading to the recognition of genes mutations in certain tumors that are associated to drug resistance [68].

Despite their utility, xenograft models may have some drawbacks. One limitation of the approach is that the immune system of the mouse is not fully functional, which may affect the response to certain therapies. Another limitation is that the stroma of mice may differ from that of humans, leading to differences in tumor growth and metastasis. Additionally, xenograft models may not accurately represent specific genotypes and lineage subtypes [69]. Moreover, these models are not suitable for evaluating treatments that rely on immune-based mechanisms or species-specific host interactions [70].

Zebrafish

Zebrafish has emerged as a promising whole organism model for evaluating signaling pathways involved in cancer as well as a model for the development of innovative therapeutics. This model organism shares significant similarities in terms of physiological and genetic characteristics with humans [71, 72]. Human hematologic malignancies, melanoma, rhabdomyosarcoma, and other solid cancers were replicated using a zebrafish model [73, 74]. Despite the incomplete development of an adaptive immune system in zebrafish larvae until 14 days after fertilization, it is possible to transplant human cancer cells into them, and these cells can survive and metastasize. This characteristic of zebrafish provides several advantages compared to standard rodent and xenograft models, as reported by [75]. The optical transparency of zebrafish embryos permits a unique opportunity to visually monitor cancer genesis and growth within the living animal [76]. In addition, zebrafish provide efficient transplantation assays using minimal tumor cells for investigating cancer self- renewal and the impact of clonal evolution on tumor initiating potential [77].

In the field of drug discovery and development, zebrafish is advantageous in that its orthotopic glioblastoma xenograft model is employed to assess the ability of drugs to penetrate the blood-brain barrier. This is due to the structural and functional similarities of the zebrafish blood brain barrier to that of humans [78].

In addition to the benefits of using zebrafish as a model system, there are also certain challenges that need to be considered. One such challenge relates to the temperature differences that are optimal for the growth and maintenance of human cells versus zebrafish. Specifically, when human cells are transplanted into zebrafish embryos, they may cease to grow when maintained at the standard temperature of 28°C used for zebrafish embryos and adult fish [79]. Furthermore, the applicability of zebrafish in cancer research is limited by the absence of certain tissue types that are present in humans, such as breast and prostate tissues. This lack of anatomical similarity reduces the utility of zebrafish models for the study of cancers affecting these specific organs [80].

Drosophila

Drosophila is a genus of the Drosophila family, members of which are often referred to as fruit flies, vinegar flies, or wine flies. The utilization of Drosophila as a model for studying human cancers has proven to be highly successful. This is due to several reasons, including its short lifespan, low genetic redundancy, rapid reproduction, economical maintenance costs, lack of ethical restrictions, and comparable genetic and functional characteristics to those of humans. It has been discovered that approximately 68% of human cancer signaling pathways are conserved in Drosophila.

In one study, researchers have already implemented Drosophila in the study of Acute Myeloid Leukemia (AML), a prevalent form of leukemia, and have successfully pinpointed the genes responsible for the disease. One of these genes, AML1, is a transcription factor that triggers myeloid differentiation and has a counterpart in the fly [81]. In addition, various studies have successfully produced Drosophila models for colorectal, lung, thyroid, and brain cancers. These models have been instrumental in conducting high-throughput screenings of FDA-approved medications. Through this process, numerous drugs were found to be effective in decreasing proliferation and reversing phenotypes. These drugs include, but are not limited to, AUH-6-96, Amsacrine, Artemisinin, curcumin, AY9944, BOT-4-one, Bouvardin, Afatinib, gefitinib, ibrutinib, bazedoxifene, afatinib, Cisplatin, and cyclophosphamide [82].

Although Drosophila is widely used as an experimental model, it has certain limitations when it comes to studying humans. One of the primary differences between the two organisms is the dissimilarity in their physiological and cancer-related mechanisms. Humans have a more intricate anatomy and physiology than Drosophila. Additionally, cancer in humans is more complex than it is in Drosophila, which means that cancer Drosophila models only provide a partial understanding of the disease in humans. These differences can lead to false positive or negative results during drug screening. Moreover, Drosophila lacks the equivalent organs of mammals, such as the liver, pancreas, spleen, thymus, kidneys, lungs, and thyroid gland [83].