1
|
De Robertis M, Signori E. Azoxymethane/Dextran Sodium Sulfate (AOM/DSS) Model of Colorectal Cancer. Methods Mol Biol 2024; 2773:51-58. [PMID: 38236535 DOI: 10.1007/978-1-0716-3714-2_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Recent progress in developing new vaccination strategies against cancer requires the production of complex and reliable animal models reflecting the complexity of the tumors with their microenvironment. Mice can be considered a good source due to low cost and ease of being genetically modified, inoculated with tumor cell lines or treated by chemicals to induce different cancers. Despite significant limitations in modeling human cancer complexity, preclinical trials conducted in mice can efficiently contribute to understand molecular mechanisms of cancer, to closely resemble and follow carcinogenesis steps impossible to study into humans, and to test new anticancer therapies. In this chapter, we generally describe the different mouse models developed for cancer vaccines' preclinical trials. A particular focus is dedicated to a chemically-induced colorectal cancer model in use in our laboratories.
Collapse
Affiliation(s)
- Mariangela De Robertis
- Department of Biosciences, Biotechnology and Environment, University of Bari 'A. Moro', Bari, Italy
| | - Emanuela Signori
- Laboratory of Molecular Pathology and Experimental Oncology, Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche, Rome, Italy.
| |
Collapse
|
2
|
Mahmoudian RA, Farshchian M, Golyan FF, Mahmoudian P, Alasti A, Moghimi V, Maftooh M, Khazaei M, Hassanian SM, Ferns GA, Mahaki H, Shahidsales S, Avan A. Preclinical tumor mouse models for studying esophageal cancer. Crit Rev Oncol Hematol 2023; 189:104068. [PMID: 37468084 DOI: 10.1016/j.critrevonc.2023.104068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/21/2023] Open
Abstract
Preclinical models are extensively employed in cancer research because they can be manipulated in terms of their environment, genome, molecular biology, organ systems, and physical activity to mimic human behavior and conditions. The progress made in in vivo cancer research has resulted in significant advancements, enabling the creation of spontaneous, metastatic, and humanized mouse models. Most recently, the remarkable and extensive developments in genetic engineering, particularly the utilization of CRISPR/Cas9, transposable elements, epigenome modifications, and liquid biopsies, have further facilitated the design and development of numerous mouse models for studying cancer. In this review, we have elucidated the production and usage of current mouse models, such as xenografts, chemical-induced models, and genetically engineered mouse models (GEMMs), for studying esophageal cancer. Additionally, we have briefly discussed various gene-editing tools that could potentially be employed in the future to create mouse models specifically for esophageal cancer research.
Collapse
Affiliation(s)
- Reihaneh Alsadat Mahmoudian
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Moein Farshchian
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Fatemeh Fardi Golyan
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvaneh Mahmoudian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Alasti
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Moghimi
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Mina Maftooh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Department of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Hanie Mahaki
- Vascular & Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq; Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
3
|
Abstract
The advent of clustered regularly interspaced short palindromic repeat (CRISPR) genome editing, coupled with advances in computing and imaging capabilities, has initiated a new era in which genetic diseases and individual disease susceptibilities are both predictable and actionable. Likewise, genes responsible for plant traits can be identified and altered quickly, transforming the pace of agricultural research and plant breeding. In this Review, we discuss the current state of CRISPR-mediated genetic manipulation in human cells, animals, and plants along with relevant successes and challenges and present a roadmap for the future of this technology.
Collapse
Affiliation(s)
- Joy Y Wang
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA.,Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Jennifer A Doudna
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA.,Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA.,Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA, USA.,Molecular Biophysics & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.,Gladstone Institutes, University of California, San Francisco, San Francisco, CA, USA.,Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| |
Collapse
|
4
|
Zhang W, Zheng X. Patient-derived xenografts or organoids in the discovery of traditional and self-assembled drug for tumor immunotherapy. Front Oncol 2023; 13:1122322. [PMID: 37081982 PMCID: PMC10110942 DOI: 10.3389/fonc.2023.1122322] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/24/2023] [Indexed: 04/22/2023] Open
Abstract
In addition to the rapid development of immune checkpoint inhibitors, there has also been a surge in the development of self-assembly immunotherapy drugs. Based on the immune target, traditional tumor immunotherapy drugs are classified into five categories, namely immune checkpoint inhibitors, direct immune modulators, adoptive cell therapy, oncolytic viruses, and cancer vaccines. Additionally, the emergence of self-assembled drugs with improved precision and environmental sensitivity offers a promising innovation approach to tumor immunotherapy. Despite rapid advances in tumor immunotherapy drug development, all candidate drugs require preclinical evaluation for safety and efficacy, and conventional evaluations are primarily conducted using two-dimensional cell lines and animal models, an approach that may be unsuitable for immunotherapy drugs. The patient-derived xenograft and organoids models, however, maintain the heterogeneity and immunity of the pathological tumor heterogeneity.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Talent Highland, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoqiang Zheng
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Xiaoqiang Zheng,
| |
Collapse
|
5
|
Targeting the "hallmarks of aging" to slow aging and treat age-related disease: fact or fiction? Mol Psychiatry 2023; 28:242-255. [PMID: 35840801 PMCID: PMC9812785 DOI: 10.1038/s41380-022-01680-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 01/09/2023]
Abstract
Aging is a major risk factor for a number of chronic diseases, including neurodegenerative and cerebrovascular disorders. Aging processes have therefore been discussed as potential targets for the development of novel and broadly effective preventatives or therapeutics for age-related diseases, including those affecting the brain. Mechanisms thought to contribute to aging have been summarized under the term the "hallmarks of aging" and include a loss of proteostasis, mitochondrial dysfunction, altered nutrient sensing, telomere attrition, genomic instability, cellular senescence, stem cell exhaustion, epigenetic alterations and altered intercellular communication. We here examine key claims about the "hallmarks of aging". Our analysis reveals important weaknesses that preclude strong and definitive conclusions concerning a possible role of these processes in shaping organismal aging rate. Significant ambiguity arises from the overreliance on lifespan as a proxy marker for aging, the use of models with unclear relevance for organismal aging, and the use of study designs that do not allow to properly estimate intervention effects on aging rate. We also discuss future research directions that should be taken to clarify if and to what extent putative aging regulators do in fact interact with aging. These include multidimensional analytical frameworks as well as designs that facilitate the proper assessment of intervention effects on aging rate.
Collapse
|
6
|
Ramírez Guerson GA, Linares Márquez P, Pascual Mathey LI. Perspectiva bioética para México, España y Estados Unidos en los modelos in vivo de inducción de daño al ADN. REVISTA LATINOAMERICANA DE BIOÉTICA 2022. [DOI: 10.18359/rlbi.5592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
En la actualidad, la investigación biomédica se ha centrado en el estudio de enfermedades como el cáncer, que causan un elevado índice de mortalidad. Existen diferentes modelos animales, empleados para generar diversos tipos de carcinogénesis; el daño directo al ADN es uno de los mecanismos más utilizados. Sin embargo, en la normatividad nacional e internacional vigente, no se señalan los aspectos bioéticos que se deben seguir para desarrollar un modelo experimental de daño al ADN. Además, no se realiza una correcta semejanza de la enfermedad. Debido a lo anterior, esta revisión analiza los avances en cuanto a normatividad que se han generado en diferentes países, comparando los estudios encontrados en Estados Unidos, México y España. La perspectiva a futuro es poder contar con guías de experimentación actualizadas, que permitan pautar las normas necesarias para el adecuado desarrollo de los modelos de investigación animal de daño al ADN y que cumplan con la regla de las 3R en la experimentación animal. Esta iniciativa se debe de realizar en conjunto entre la Organización Mundial de la Salud y los organismos especializados en manejo y cuidado de animales de laboratorio en los ámbitos nacional e internacional.
Collapse
|
7
|
Dozzo A, Galvin A, Shin JW, Scalia S, O'Driscoll CM, Ryan KB. Modelling acute myeloid leukemia (AML): What's new? A transition from the classical to the modern. Drug Deliv Transl Res 2022:10.1007/s13346-022-01189-4. [PMID: 35930221 DOI: 10.1007/s13346-022-01189-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 11/24/2022]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous malignancy affecting myeloid cells in the bone marrow (BM) but can spread giving rise to impaired hematopoiesis. AML incidence increases with age and is associated with poor prognostic outcomes. There has been a disconnect between the success of novel drug compounds observed in preclinical studies of hematological malignancy and less than exceptional therapeutic responses in clinical trials. This review aims to provide a state-of-the-art overview on the different preclinical models of AML available to expand insights into disease pathology and as preclinical screening tools. Deciphering the complex physiological and pathological processes and developing predictive preclinical models are key to understanding disease progression and fundamental in the development and testing of new effective drug treatments. Standard scaffold-free suspension models fail to recapitulate the complex environment where AML occurs. To this end, we review advances in scaffold/matrix-based 3D models and outline the most recent advances in on-chip technology. We also provide an overview of clinically relevant animal models and review the expanding use of patient-derived samples, which offer the prospect to create more "patient specific" screening tools either in the guise of 3D matrix models, microphysiological "organ-on-chip" tools or xenograft models and discuss representative examples.
Collapse
Affiliation(s)
| | - Aoife Galvin
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Jae-Won Shin
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, 909 S. Wolcott Ave, Chicago, IL, 5091 COMRB, USA
| | - Santo Scalia
- Università degli Studi di Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Caitriona M O'Driscoll
- School of Pharmacy, University College Cork, Cork, Ireland.,SSPC Centre for Pharmaceutical Research, School of Pharmacy, University College Cork, Cork, Ireland
| | - Katie B Ryan
- School of Pharmacy, University College Cork, Cork, Ireland. .,SSPC Centre for Pharmaceutical Research, School of Pharmacy, University College Cork, Cork, Ireland.
| |
Collapse
|
8
|
Connolly KA, Fitzgerald B, Damo M, Joshi NS. Novel Mouse Models for Cancer Immunology. ANNUAL REVIEW OF CANCER BIOLOGY 2022; 6:269-291. [PMID: 36875867 PMCID: PMC9979244 DOI: 10.1146/annurev-cancerbio-070620-105523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mouse models for the study of cancer immunology provide excellent systems in which to test biological mechanisms of the immune response against cancer. Historically, these models have been designed to have different strengths based on the current major research questions at the time. As such, many mouse models of immunology used today were not originally developed to study questions currently plaguing the relatively new field of cancer immunology, but instead have been adapted for such purposes. In this review, we discuss various mouse model of cancer immunology in a historical context as a means to provide a fuller perspective of each model's strengths. From this outlook, we discuss the current state of the art and strategies for tackling future modeling challenges.
Collapse
Affiliation(s)
- Kelli A. Connolly
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Brittany Fitzgerald
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Martina Damo
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Nikhil S. Joshi
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| |
Collapse
|
9
|
Guiren Fritah H, Rovelli R, Lai-Lai Chiang C, Kandalaft LE. The current clinical landscape of personalized cancer vaccines. Cancer Treat Rev 2022; 106:102383. [DOI: 10.1016/j.ctrv.2022.102383] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/20/2022] [Indexed: 12/11/2022]
|
10
|
Arnal-Estapé A, Foggetti G, Starrett JH, Nguyen DX, Politi K. Preclinical Models for the Study of Lung Cancer Pathogenesis and Therapy Development. Cold Spring Harb Perspect Med 2021; 11:a037820. [PMID: 34518338 PMCID: PMC8634791 DOI: 10.1101/cshperspect.a037820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Experimental preclinical models have been a cornerstone of lung cancer translational research. Work in these model systems has provided insights into the biology of lung cancer subtypes and their origins, contributed to our understanding of the mechanisms that underlie tumor progression, and revealed new therapeutic vulnerabilities. Initially patient-derived lung cancer cell lines were the main preclinical models available. The landscape is very different now with numerous preclinical models for research each with unique characteristics. These include genetically engineered mouse models (GEMMs), patient-derived xenografts (PDXs) and three-dimensional culture systems ("organoid" cultures). Here we review the development and applications of these models and describe their contributions to lung cancer research.
Collapse
Affiliation(s)
- Anna Arnal-Estapé
- Department of Pathology
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | | | | | - Don X Nguyen
- Department of Pathology
- Department of Internal Medicine (Section of Medical Oncology)
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Katerina Politi
- Department of Pathology
- Department of Internal Medicine (Section of Medical Oncology)
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| |
Collapse
|
11
|
Mahmoudian RA, Farshchian M, Abbaszadegan MR. Genetically engineered mouse models of esophageal cancer. Exp Cell Res 2021; 406:112757. [PMID: 34331909 DOI: 10.1016/j.yexcr.2021.112757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/10/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Esophageal cancer is the most common cause of cancer-related death worldwide with a diverse geographical distribution, poor prognosis, and diagnosis in advanced stages of the disease. Identification of the mechanisms involved in esophageal cancer development is evaluative to improve outcomes for patients. Genetically engineered mouse models (GEMMs) of cancer provide the physiologic, molecular, and histologic features of the human tumors to determine the pathogenesis and treatments for cancer, hence exhibiting a source of tremendous potential for oncology research. The advancement of cancer modeling in mice has improved to the extent that researchers can observe and manipulate the disease process in a specific manner. Despite the significant differences between mice and humans, mice can be great models for human oncology researches due to similarities between them at the molecular and physiological levels. Due to most of the existing esophageal cancer GEMMs do not propose an ideal system for pathogenesis of the disease, genetic risks, and microenvironment exposure, so identification of challenges in GEM modeling and well-developed technologies are required to obtain the most value for patients. In this review, we describe the biology of human and mouse, followed by the exciting esophageal cancer mouse models with a discussion of applicability and challenges of these models for generating new GEMMs in future studies.
Collapse
Affiliation(s)
| | - Moein Farshchian
- Stem Cell and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi, Mashhad, Iran.
| | | |
Collapse
|
12
|
Sajjad H, Imtiaz S, Noor T, Siddiqui YH, Sajjad A, Zia M. Cancer models in preclinical research: A chronicle review of advancement in effective cancer research. Animal Model Exp Med 2021; 4:87-103. [PMID: 34179717 PMCID: PMC8212826 DOI: 10.1002/ame2.12165] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/04/2021] [Indexed: 12/15/2022] Open
Abstract
Cancer is a major stress for public well-being and is the most dreadful disease. The models used in the discovery of cancer treatment are continuously changing and extending toward advanced preclinical studies. Cancer models are either naturally existing or artificially prepared experimental systems that show similar features with human tumors though the heterogeneous nature of the tumor is very familiar. The choice of the most fitting model to best reflect the given tumor system is one of the real difficulties for cancer examination. Therefore, vast studies have been conducted on the cancer models for developing a better understanding of cancer invasion, progression, and early detection. These models give an insight into cancer etiology, molecular basis, host tumor interaction, the role of microenvironment, and tumor heterogeneity in tumor metastasis. These models are also used to predict novel cancer markers, targeted therapies, and are extremely helpful in drug development. In this review, the potential of cancer models to be used as a platform for drug screening and therapeutic discoveries are highlighted. Although none of the cancer models is regarded as ideal because each is associated with essential caveats that restraint its application yet by bridging the gap between preliminary cancer research and translational medicine. However, they promise a brighter future for cancer treatment.
Collapse
Affiliation(s)
- Humna Sajjad
- Department of BiotechnologyQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Saiqa Imtiaz
- Department of BiotechnologyQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Tayyaba Noor
- Department of BiotechnologyQuaid‐i‐Azam UniversityIslamabadPakistan
| | | | - Anila Sajjad
- Department of BiotechnologyQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Muhammad Zia
- Department of BiotechnologyQuaid‐i‐Azam UniversityIslamabadPakistan
| |
Collapse
|
13
|
Ruiz-Espigares J, Nieto D, Moroni L, Jiménez G, Marchal JA. Evolution of Metastasis Study Models toward Metastasis-On-A-Chip: The Ultimate Model? SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006009. [PMID: 33705602 DOI: 10.1002/smll.202006009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/05/2020] [Indexed: 06/12/2023]
Abstract
For decades, several attempts have been made to obtain a mimetic model for the study of metastasis, the reason of most of deaths caused by cancer, in order to solve the unknown phenomena surrounding this disease. To better understand this cellular dissemination process, more realistic models are needed that are capable of faithfully recreating the entire and essential tumor microenvironment (TME). Thus, new tools known as tumor-on-a-chip and metastasis-on-a-chip have been recently proposed. These tools incorporate microfluidic systems and small culture chambers where TME can be faithfully modeled thanks to 3D bioprinting. In this work, a literature review has been developed about the different phases of metastasis, the remaining unknowns and the use of new models to study this disease. The aim is to provide a global vision of the current panorama and the great potential that these systems have for in vitro translational research on the molecular basis of the pathology. In addition, these models will allow progress toward a personalized medicine, generating chips from patient samples that mimic the original tumor and the metastatic process to perform a precise pharmacological screening by establishing the most appropriate treatment protocol.
Collapse
Affiliation(s)
- Jesús Ruiz-Espigares
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, E-18016, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18016, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, E-18016, Spain
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, Granada, E-18071, Spain
| | - Daniel Nieto
- Photonics4life Research Group, Applied Physics Department, Faculty of Physics, University of Santiago de Compostela, Santiago de Compostela, 15705, Spain
- Complex Tissue Regeneration Department, MERLN Institute for Technology Inspired Regenerative Medicine, Universiteitssingel 40, Maastricht, 6229ER, The Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration Department, MERLN Institute for Technology Inspired Regenerative Medicine, Universiteitssingel 40, Maastricht, 6229ER, The Netherlands
| | - Gema Jiménez
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, E-18016, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18016, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, E-18016, Spain
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, Granada, E-18071, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, E-18016, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18016, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, E-18016, Spain
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, Granada, E-18071, Spain
| |
Collapse
|
14
|
Redler G, Pearson E, Liu X, Gertsenshteyn I, Epel B, Pelizzari C, Aydogan B, Weichselbaum R, Halpern HJ, Wiersma RD. Small Animal IMRT Using 3D-Printed Compensators. Int J Radiat Oncol Biol Phys 2020; 110:551-565. [PMID: 33373659 DOI: 10.1016/j.ijrobp.2020.12.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 11/17/2020] [Accepted: 12/13/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE Preclinical radiation replicating clinical intensity modulated radiation therapy (IMRT) techniques can provide data translatable to clinical practice. For this work, treatment plans were created for oxygen-guided dose-painting in small animals using inverse-planned IMRT. Spatially varying beam intensities were achieved using 3-dimensional (3D)-printed compensators. METHODS AND MATERIALS Optimized beam fluence from arbitrary gantry angles was determined using a verified model of the XRAD225Cx treatment beam. Compensators were 3D-printed with varied thickness to provide desired attenuation using copper/polylactic-acid. Spatial resolution capabilities were investigated using printed test-patterns. Following American Association of Physicists in Medicine TG119, a 5-beam IMRT plan was created for a miniaturized (∼1/8th scale) C-shape target. Electron paramagnetic resonance imaging of murine tumor oxygenation guided simultaneous integrated boost (SIB) plans conformally treating tumor to a base dose (Rx1) with boost (Rx2) based on tumor oxygenation. The 3D-printed compensator intensity modulation accuracy and precision was evaluated by individually delivering each field to a phantom containing radiochromic film and subsequent per-field gamma analysis. The methodology was validated end-to-end with composite delivery (incorporating 3D-printed tungsten/polylactic-acid beam trimmers to reduce out-of-field leakage) of the oxygen-guided SIB plan to a phantom containing film and subsequent gamma analysis. RESULTS Resolution test-patterns demonstrate practical printer resolution of ∼0.7 mm, corresponding to 1.0 mm bixels at the isocenter. The miniaturized C-shape plan provides planning target volume coverage (V95% = 95%) with organ sparing (organs at risk Dmax < 50%). The SIB plan to hypoxic tumor demonstrates the utility of this approach (hypoxic tumor V95%,Rx2 = 91.6%, normoxic tumor V95%,Rx1 = 95.7%, normal tissue V100%,Rx1 = 7.1%). The more challenging SIB plan to boost the normoxic tumor rim achieved normoxic tumor V95%,Rx2 = 90.9%, hypoxic tumor V95%,Rx1 = 62.7%, and normal tissue V100%,Rx2 = 5.3%. Average per-field gamma passing rates using 3%/1.0 mm, 3%/0.7 mm, and 3%/0.5 mm criteria were 98.8% ± 2.8%, 96.6% ± 4.1%, and 90.6% ± 5.9%, respectively. Composite delivery of the hypoxia boost plan and gamma analysis (3%/1 mm) gave passing results of 95.3% and 98.1% for the 2 measured orthogonal dose planes. CONCLUSIONS This simple and cost-effective approach using 3D-printed compensators for small-animal IMRT provides a methodology enabling preclinical studies that can be readily translated into the clinic. The presented oxygen-guided dose-painting demonstrates that this methodology will facilitate studies driving much needed biologic personalization of radiation therapy for improvements in patient outcomes.
Collapse
Affiliation(s)
- Gage Redler
- Moffitt Cancer Center, Department of Radiation Oncology, Tampa, Florida.
| | - Erik Pearson
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Xinmin Liu
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Inna Gertsenshteyn
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Boris Epel
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Charles Pelizzari
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Bulent Aydogan
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Ralph Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Howard J Halpern
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Rodney D Wiersma
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
15
|
Guerin MV, Finisguerra V, Van den Eynde BJ, Bercovici N, Trautmann A. Preclinical murine tumor models: a structural and functional perspective. eLife 2020; 9:e50740. [PMID: 31990272 PMCID: PMC6986875 DOI: 10.7554/elife.50740] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/06/2020] [Indexed: 12/14/2022] Open
Abstract
The goal of this review is to pinpoint the specific features, including the weaknesses, of various tumor models, and to discuss the reasons why treatments that are efficient in murine tumor models often do not work in clinics. In a detailed comparison of transplanted and spontaneous tumor models, we focus on structure-function relationships in the tumor microenvironment. For instance, the architecture of the vascular tree, which depends on whether tumor cells have gone through epithelial-mesenchymal transition, is determinant for the extension of the spontaneous necrosis, and for the intratumoral localization of the immune infiltrate. Another key point is the model-dependent abundance of TGFβ in the tumor, which controls the variable susceptibility of different tumor models to treatments. Grounded in a historical perspective, this review provides a rationale for checking factors that will be key for the transition between preclinical murine models and clinical applications.
Collapse
Affiliation(s)
- Marion V Guerin
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, F-75014ParisFrance
| | - Veronica Finisguerra
- Ludwig Institute for Cancer Research, de Duve Institute WELBIOUCLouvainBrusselsBelgium
| | | | - Nadege Bercovici
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, F-75014ParisFrance
| | - Alain Trautmann
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, F-75014ParisFrance
| |
Collapse
|
16
|
Mendes N, Dias Carvalho P, Martins F, Mendonça S, Malheiro AR, Ribeiro A, Carvalho J, Velho S. Animal Models to Study Cancer and Its Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:389-401. [PMID: 32130710 DOI: 10.1007/978-3-030-34025-4_20] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cancers are complex tissues composed by genetically altered cancer cells and stromal elements such as inflammatory/immune cells, fibroblasts, endothelial cells and pericytes, neuronal cells, and a non-cellular component, the extracellular matrix. The complex network of interactions and crosstalk established between cancer cells and the supportig cellular and non-cellular components of the microenvironment are of extreme importance for tumor initiation and progression, strongly impacting the course and the outcome of the disease. Therefore, a better understanding of the tumorigenic processes implies the combined study of the cancer cell and the biologic, chemical and mechanic constituents of the tumor microenvironment, as their concerted action plays a major role in the carcinogenic pathway and is a key determinant of the efficacy of anti-cancer treatments. The use of animal models (e.g. Mouse, Zebrafish and Drosophila) to study cancer has greatly impacted our understanding of the processes governing initiation, progression and metastasis and allowed the discovery and pre-clinical validation of novel cancer treatments as it allows to recreate tumor development in a more pathophysiologic environment.
Collapse
Affiliation(s)
- N Mendes
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal.
- IPATIMUP, Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal.
| | - P Dias Carvalho
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IPATIMUP, Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal
| | - F Martins
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IPATIMUP, Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal
| | - S Mendonça
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IPATIMUP, Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal
| | - A R Malheiro
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IBMC, Instituto de Biologia Molecular e Celular da Universidade do Porto, Porto, Portugal
| | - A Ribeiro
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IPATIMUP, Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal
| | - J Carvalho
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IPATIMUP, Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal
| | - S Velho
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal.
- IPATIMUP, Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal.
| |
Collapse
|
17
|
Seidlitz T, Chen YT, Uhlemann H, Schölch S, Kochall S, Merker SR, Klimova A, Hennig A, Schweitzer C, Pape K, Baretton GB, Welsch T, Aust DE, Weitz J, Koo BK, Stange DE. Mouse Models of Human Gastric Cancer Subtypes With Stomach-Specific CreERT2-Mediated Pathway Alterations. Gastroenterology 2019; 157:1599-1614.e2. [PMID: 31585123 PMCID: PMC6902245 DOI: 10.1053/j.gastro.2019.09.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 09/02/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Patterns of genetic alterations characterize different molecular subtypes of human gastric cancer. We aimed to establish mouse models of these subtypes. METHODS We searched databases to identify genes with unique expression in the stomach epithelium, resulting in the identification of Anxa10. We generated mice with tamoxifen-inducible Cre recombinase (CreERT2) in the Anxa10 gene locus. We created 3 mouse models with alterations in pathways that characterize the chromosomal instability (CIN) and the genomically stable (GS) subtypes of human gastric cancer: Anxa10-CreERT2;KrasG12D/+;Tp53R172H/+;Smad4fl/f (CIN mice), Anxa10-CreERT2;Cdh1fl/fl;KrasG12D/+;Smad4fl/fl (GS-TGBF mice), and Anxa10-CreERT2;Cdh1fl/fl;KrasG12D/+;Apcfl/fl (GS-Wnt mice). We analyzed tumors that developed in these mice by histology for cell types and metastatic potential. We derived organoids from the tumors and tested their response to chemotherapeutic agents and the epithelial growth factor receptor signaling pathway inhibitor trametinib. RESULTS The gastric tumors from the CIN mice had an invasive phenotype and formed liver and lung metastases. The tumor cells had a glandular morphology, similar to human intestinal-type gastric cancer. The gastric tumors from the GS-TGFB mice were poorly differentiated with diffuse morphology and signet ring cells, resembling human diffuse-type gastric cancer. Cells from these tumors were invasive, and mice developed peritoneal carcinomatosis and lung metastases. GS-Wnt mice developed adenomatous tooth-like gastric cancer. Organoids derived from tumors of GS-TGBF and GS-Wnt mice were more resistant to docetaxel, whereas organoids from the CIN tumors were more resistant to trametinib. CONCLUSIONS Using a stomach-specific CreERT2 system, we created mice that develop tumors with morphologic similarities to subtypes of human gastric cancer. These tumors have different patterns of local growth, metastasis, and response to therapeutic agents. They can be used to study different subtypes of human gastric cancer.
Collapse
Affiliation(s)
- Therese Seidlitz
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Yi-Ting Chen
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan,Department of Pathology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Heike Uhlemann
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sebastian Schölch
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany,German Cancer Consortium (DKTK), Dresden, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Susan Kochall
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sebastian R. Merker
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anna Klimova
- Institute for Medical Informatics and Biometry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,Core Unit for Data Management and Analytics (CDMA), National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Alexander Hennig
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,National Center for Tumor Diseases, Dresden, Germany
| | - Christine Schweitzer
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kristin Pape
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Gustavo B. Baretton
- German Cancer Consortium (DKTK), Dresden, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany,Core Unit for Molecular Tumour Diagnostics, National Center for Tumor Diseases (NCT), Dresden, Germany,Institute of Pathology and Tumour and Normal Tissue Bank of the University Cancer Center, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany,National Center for Tumor Diseases, Dresden, Germany
| | - Thilo Welsch
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Daniela E. Aust
- German Cancer Consortium (DKTK), Dresden, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany,Core Unit for Molecular Tumour Diagnostics, National Center for Tumor Diseases (NCT), Dresden, Germany,Institute of Pathology and Tumour and Normal Tissue Bank of the University Cancer Center, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany,National Center for Tumor Diseases, Dresden, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,German Cancer Consortium (DKTK), Dresden, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany,National Center for Tumor Diseases, Dresden, Germany
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Daniel E. Stange
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,German Cancer Consortium (DKTK), Dresden, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany,National Center for Tumor Diseases, Dresden, Germany,Reprint requests Address requests for reprints to: Daniel E. Strange, MD, PhD, Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| |
Collapse
|
18
|
Woods K, Nguyen D, Neph R, Ruan D, O'Connor D, Sheng K. A sparse orthogonal collimator for small animal intensity-modulated radiation therapy part I: Planning system development and commissioning. Med Phys 2019; 46:5703-5713. [PMID: 31621920 DOI: 10.1002/mp.13872] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 12/18/2022] Open
Abstract
PURPOSE To achieve more translatable preclinical research results, small animal irradiation needs to more closely simulate human radiotherapy. Although the clinical gold standard is intensity-modulated radiation therapy (IMRT), the direct translation of this method for small animals is impractical. In this study we describe the treatment planning system for a novel dose modulation device to address this challenge. METHODS Using delineated target and avoidance structures, a rectangular aperture optimization (RAO) problem was formulated to penalize deviations from a desired dose distribution and limit the number of selected rectangular apertures. RAO was used to create IMRT plans with highly concave targets in the mouse brain, and the plan quality was compared to that using a hypothetical miniaturized multileaf collimator (MLC). RAO plans were also created for a realistic application of mouse whole liver irradiation and for a highly complex two-dimensional (2D) dose distribution as a proof-of-principle. Beam commissioning data, including output and off-axis factors and percent depth dose (PDD) curves, were acquired for our small animal irradiation system and incorporated into the treatment planning system. A plan post-processing step was implemented for aperture size-specific dose recalculation and aperture weighting reoptimization. RESULTS The first RAO test case achieved highly conformal doses to concave targets in the brain, with substantially better dose gradient, conformity, and target dose homogeneity than the hypothetical miniaturized MLC plans. In the second test case, a highly conformal dose to the liver was achieved with significant sparing of the kidneys. RAO also successfully replicated a complex 2D dose distribution with three prescription dose levels. Energy spectra for field sizes 1 to 20 mm were calculated to match the measured PDD curves, with maximum and mean dose deviations of 4.47 ± 0.30% and 1.71 ± 0.18%. The final reoptimization of aperture weightings for the complex RAO test plan was able to reduce the maximum and mean dose deviations between the optimized and recalculated dose distributions from 10.3% to 6.6% and 4.0% to 2.8%, respectively. CONCLUSIONS Using the advanced optimization techniques, complex IMRT plans were achieved using a simple dose modulation device. Beam commissioning data were incorporated into the treatment planning process to more accurately predict the resulting dose distribution. This platform substantially reduces the gap in treatment plan quality between clinical and preclinical radiotherapy, potentially increasing the value and flexibility of small animal studies.
Collapse
Affiliation(s)
- Kaley Woods
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Dan Nguyen
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Ryan Neph
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Dan Ruan
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Daniel O'Connor
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Ke Sheng
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
19
|
Ireson CR, Alavijeh MS, Palmer AM, Fowler ER, Jones HJ. The role of mouse tumour models in the discovery and development of anticancer drugs. Br J Cancer 2019; 121:101-108. [PMID: 31231121 PMCID: PMC6738037 DOI: 10.1038/s41416-019-0495-5] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 05/09/2019] [Accepted: 05/16/2019] [Indexed: 02/06/2023] Open
Abstract
Our understanding of cancer biology has increased substantially over the past 30 years. Despite this, and an increasing pharmaceutical company expenditure on research and development, the approval of novel oncology drugs during the past decade continues to be modest. In addition, the attrition of agents during clinical development remains high. This attrition can be attributed, at least in part, to the clinical development being underpinned by the demonstration of predictable efficacy in experimental models of human tumours. This review will focus on the range of models available for the discovery and development of anticancer drugs, from traditional subcutaneous injection of tumour cell lines to mice genetically engineered to spontaneously give rise to tumours. It will consider the best time to use the models, along with practical applications and shortcomings. Finally, and most importantly, it will describe how these models reflect the underlying cancer biology and how well they predict efficacy in the clinic. Developing a line of sight to the clinic early in a drug discovery project provides clear benefit, as it helps to guide the selection of appropriate preclinical models and facilitates the investigation of relevant biomarkers.
Collapse
Affiliation(s)
| | - Mo S Alavijeh
- Pharmidex Pharmaceutical Services, 14 Hanover Street, London, W1S 1YH, UK
| | - Alan M Palmer
- Reading School of Pharmacy, Whiteknights, Reading, RG6 6A, UK
| | - Emily R Fowler
- Pharmidex Pharmaceutical Services, 14 Hanover Street, London, W1S 1YH, UK.,Wellcome Centre for Cell Biology and Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, EH9 3BF, Scotland, UK
| | | |
Collapse
|
20
|
Li Q, Qin Z, Wang Q, Xu T, Yang Y, He Z. Applications of Genome Editing Technology in Animal Disease Modeling and Gene Therapy. Comput Struct Biotechnol J 2019; 17:689-698. [PMID: 31303973 PMCID: PMC6603303 DOI: 10.1016/j.csbj.2019.05.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/24/2019] [Accepted: 05/26/2019] [Indexed: 02/05/2023] Open
Abstract
Genome editing technology is a technique for targeted genetic modifications, enabling the knockout and addition of specific DNA fragments. This technology has been widely used in various types of biomedical research, clinics and agriculture. In terms of disease research, constructing appropriate animal models is necessary. Combining reproductive technology with genome editing, many animal disease models have been generated for basic and clinical research. In addition, precisely targeted modifications allow genome editing to flourish in the field of gene therapy. Many mutations refractory to traditional gene therapy could be permanently corrected at the DNA level. Thus, genome editing is undoubtedly a promising technology for gene therapy. In this review, we mainly introduce the applications of genome editing in constructing animal disease models and gene therapies, as well as its future prospects and challenges.
Collapse
Affiliation(s)
- Qian Li
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Zhou Qin
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Qingnan Wang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Ting Xu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Zhiyao He
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| |
Collapse
|
21
|
Chen M, Pandolfi PP. Preclinical and Coclinical Studies in Prostate Cancer. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a030544. [PMID: 29038335 DOI: 10.1101/cshperspect.a030544] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Men who develop metastatic castration-resistant prostate cancer (mCRPC) will invariably succumb to their disease. Thus there remains a pressing need for preclinical testing of new drugs and drug combinations for late-stage prostate cancer (PCa). Insights from the mCRPC genomic landscape have revealed that, in addition to sustained androgen receptor (AR) signaling, there are other actionable molecular alterations and distinct molecular subclasses of PCa; however, the rate at which this knowledge translates into patient care via current preclinical testing is painfully slow and inefficient. Here, we will highlight the issues involved and discuss a new translational platform, "the co-clinical trial project," to expedite current preclinical studies and optimize clinical trial and experimental drug testing. With this platform, in vivo preclinical and early clinical studies are closely aligned, enabling in vivo testing of drugs using genetically engineered mouse models (GEMMs) in defined genetic contexts to personalize individual therapies. We will discuss the principles and essential components of this novel paradigm, representative success stories and future therapeutic options for mCRPC that should be explored.
Collapse
Affiliation(s)
- Ming Chen
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| |
Collapse
|
22
|
Liang P, Zhang X, Chen Y, Huang J. Developmental history and application of CRISPR in human disease. J Gene Med 2018. [PMID: 28623876 DOI: 10.1002/jgm.2963] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Genome-editing tools are programmable artificial nucleases, mainly including zinc-finger nucleases, transcription activator-like effector nucleases and clustered regularly interspaced short palindromic repeat (CRISPR). By recognizing and cleaving specific DNA sequences, genome-editing tools make it possible to generate site-specific DNA double-strand breaks (DSBs) in the genome. DSBs will then be repaired by either error-prone nonhomologous end joining or high-fidelity homologous recombination mechanisms. Through these two different mechanisms, endogenous genes can be knocked out or precisely repaired/modified. Rapid developments in genome-editing tools, especially CRISPR, have revolutionized human disease models generation, for example, various zebrafish, mouse, rat, pig, monkey and human cell lines have been constructed. Here, we review the developmental history of CRISPR and its application in studies of human diseases. In addition, we also briefly discussed the therapeutic application of CRISPR in the near future.
Collapse
Affiliation(s)
- Puping Liang
- Key Laboratory of Gene Engineering of the Ministry of Education and State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Reproductive Medicine of G uangdong Province, The Third Affiliated Hospital, Guangzhou Medical University and School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiya Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education and State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yuxi Chen
- Key Laboratory of Gene Engineering of the Ministry of Education and State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Junjiu Huang
- Key Laboratory of Gene Engineering of the Ministry of Education and State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Reproductive Medicine of G uangdong Province, The Third Affiliated Hospital, Guangzhou Medical University and School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Affiliation(s)
- Andrea Ventura
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lukas E. Dow
- Department of Medicine, Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
24
|
Zhang Z, Christin JR, Wang C, Ge K, Oktay MH, Guo W. Mammary-Stem-Cell-Based Somatic Mouse Models Reveal Breast Cancer Drivers Causing Cell Fate Dysregulation. Cell Rep 2018; 16:3146-3156. [PMID: 27653681 DOI: 10.1016/j.celrep.2016.08.048] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 07/27/2016] [Accepted: 08/16/2016] [Indexed: 01/23/2023] Open
Abstract
Cancer genomics has provided an unprecedented opportunity for understanding genetic causes of human cancer. However, distinguishing which mutations are functionally relevant to cancer pathogenesis remains a major challenge. We describe here a mammary stem cell (MaSC) organoid-based approach for rapid generation of somatic genetically engineered mouse models (GEMMs). By using RNAi and CRISPR-mediated genome engineering in MaSC-GEMMs, we have discovered that inactivation of Ptpn22 or Mll3, two genes mutated in human breast cancer, greatly accelerated PI3K-driven mammary tumorigenesis. Using these tumor models, we have also identified genetic alterations promoting tumor metastasis and causing resistance to PI3K-targeted therapy. Both Ptpn22 and Mll3 inactivation resulted in disruption of mammary gland differentiation and an increase in stem cell activity. Mechanistically, Mll3 deletion enhanced stem cell activity through activation of the HIF pathway. Thus, our study has established a robust in vivo platform for functional cancer genomics and has discovered functional breast cancer mutations.
Collapse
Affiliation(s)
- Zheng Zhang
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - John R Christin
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Chunhui Wang
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kai Ge
- Adipocyte Biology and Gene Regulation Section, Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20814, USA
| | - Maja H Oktay
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10467, USA; Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Wenjun Guo
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
25
|
The fourth annual BRDS on genome editing and silencing for precision medicines. Drug Deliv Transl Res 2017; 8:266-272. [PMID: 29209906 DOI: 10.1007/s13346-017-0457-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Precision medicine is promising for treating human diseases, as it focuses on tailoring drugs to a patient's genes, environment, and lifestyle. The need for personalized medicines has opened the doors for turning nucleic acids into therapeutics. Although gene therapy has the potential to treat and cure genetic and acquired diseases, it needs to overcome certain obstacles before creating the overall prescription drugs. Recent advancement in the life science has helped to understand the effective manipulation and delivery of genome-engineering tools better. The use of sequence-specific nucleases allows genetic changes in human cells to be easily made with higher efficiency and precision than before. Nanotechnology has made rapid advancement in the field of drug delivery, but the delivery of nucleic acids presents unique challenges. Also, designing efficient and short time-consuming genome-editing tools with negligible off-target effects are in high demand for precision medicine. In the fourth annual Biopharmaceutical Research and Development Symposium (BRDS) held at the University of Nebraska Medical Center (UNMC) on September 7-8, 2017, we covered different facets of developing tools for precision medicine for therapeutic and diagnosis of genetic disorders.
Collapse
|
26
|
Imran A, Qamar HY, Ali Q, Naeem H, Riaz M, Amin S, Kanwal N, Ali F, Sabar MF, Nasir IA. Role of Molecular Biology in Cancer Treatment: A Review Article. IRANIAN JOURNAL OF PUBLIC HEALTH 2017; 46:1475-1485. [PMID: 29167765 PMCID: PMC5696686 DOI: pmid/29167765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Background: Cancer is a genetic disease and mainly arises due to a number of reasons include activation of onco-genes, malfunction of tumor suppressor genes or mutagenesis due to external factors. Methods: This article was written from the data collected from PubMed, Nature, Science Direct, Springer and Elsevier groups of journals. Results: Oncogenes are deregulated form of normal proto-oncogenes required for cell division, differentiation and regulation. The conversion of proto-oncogene to oncogene is caused due to translocation, rearrangement of chromosomes or mutation in gene due to addition, deletion, duplication or viral infection. These oncogenes are targeted by drugs or RNAi system to prevent proliferation of cancerous cells. There have been developed different techniques of molecular biology used to diagnose and treat cancer, including retroviral therapy, silencing of oncogenes and mutations in tumor suppressor genes. Conclusion: Among all the techniques used, RNAi, zinc finger nucleases and CRISPR hold a brighter future towards creating a Cancer Free World.
Collapse
Affiliation(s)
- Aman Imran
- Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Hafiza Yasara Qamar
- Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Qurban Ali
- Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan.,Institute of Molecular Biology and Biotechnology, University of Lahore, Lahore, Pakistan
| | - Hafsa Naeem
- Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Mariam Riaz
- Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Saima Amin
- Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Naila Kanwal
- Dept. of Plant Breeding and Genetics, University of Agriculture, Faisalabad, Pakistan
| | - Fawad Ali
- Dept. of Plant Breeding and Genetics, University of Agriculture, Faisalabad, Pakistan.,Southern Cross Plant Science, Southern Cross University, Lismore, Australia
| | | | - Idrees Ahmad Nasir
- Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
27
|
Development of zebrafish medulloblastoma-like PNET model by TALEN-mediated somatic gene inactivation. Oncotarget 2017; 8:55280-55297. [PMID: 28903419 PMCID: PMC5589658 DOI: 10.18632/oncotarget.19424] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 07/11/2017] [Indexed: 01/09/2023] Open
Abstract
Genetically engineered animal tumor models have traditionally been generated by the gain of single or multiple oncogenes or the loss of tumor suppressor genes; however, the development of live animal models has been difficult given that cancer phenotypes are generally induced by somatic mutation rather than by germline genetic inactivation. In this study, we developed somatically mutated tumor models using TALEN-mediated somatic gene inactivation of cdkn2a/b or rb1 tumor suppressor genes in zebrafish. One-cell stage injection of cdkn2a/b-TALEN mRNA resulted in malignant peripheral nerve sheath tumors with high frequency (about 39%) and early onset (about 35 weeks of age) in F0 tp53e7/e7 mutant zebrafish. Injection of rb1-TALEN mRNA also led to the formation of brain tumors at high frequency (58%, 31 weeks of age) in F0 tp53e7/e7 mutant zebrafish. Analysis of each tumor induced by somatic inactivation showed that the targeted genes had bi-allelic mutations. Tumors induced by rb1 somatic inactivation were characterized as medulloblastoma-like primitive neuroectodermal tumors based on incidence location, histopathological features, and immunohistochemical tests. In addition, 3' mRNA Quanti-Seq analysis showed differential activation of genes involved in cell cycle, DNA replication, and protein synthesis; especially, genes involved in neuronal development were up-regulated.
Collapse
|
28
|
Rigoutsos I, Lee SK, Nam SY, Anfossi S, Pasculli B, Pichler M, Jing Y, Rodriguez-Aguayo C, Telonis AG, Rossi S, Ivan C, Catela Ivkovic T, Fabris L, Clark PM, Ling H, Shimizu M, Redis RS, Shah MY, Zhang X, Okugawa Y, Jung EJ, Tsirigos A, Huang L, Ferdin J, Gafà R, Spizzo R, Nicoloso MS, Paranjape AN, Shariati M, Tiron A, Yeh JJ, Teruel-Montoya R, Xiao L, Melo SA, Menter D, Jiang ZQ, Flores ER, Negrini M, Goel A, Bar-Eli M, Mani SA, Liu CG, Lopez-Berestein G, Berindan-Neagoe I, Esteller M, Kopetz S, Lanza G, Calin GA. N-BLR, a primate-specific non-coding transcript leads to colorectal cancer invasion and migration. Genome Biol 2017; 18:98. [PMID: 28535802 PMCID: PMC5442648 DOI: 10.1186/s13059-017-1224-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/26/2017] [Indexed: 12/13/2022] Open
Abstract
Background Non-coding RNAs have been drawing increasing attention in recent years as functional data suggest that they play important roles in key cellular processes. N-BLR is a primate-specific long non-coding RNA that modulates the epithelial-to-mesenchymal transition, facilitates cell migration, and increases colorectal cancer invasion. Results We performed multivariate analyses of data from two independent cohorts of colorectal cancer patients and show that the abundance of N-BLR is associated with tumor stage, invasion potential, and overall patient survival. Through in vitro and in vivo experiments we found that N-BLR facilitates migration primarily via crosstalk with E-cadherin and ZEB1. We showed that this crosstalk is mediated by a pyknon, a short ~20 nucleotide-long DNA motif contained in the N-BLR transcript and is targeted by members of the miR-200 family. In light of these findings, we used a microarray to investigate the expression patterns of other pyknon-containing genomic loci. We found multiple such loci that are differentially transcribed between healthy and diseased tissues in colorectal cancer and chronic lymphocytic leukemia. Moreover, we identified several new loci whose expression correlates with the colorectal cancer patients’ overall survival. Conclusions The primate-specific N-BLR is a novel molecular contributor to the complex mechanisms that underlie metastasis in colorectal cancer and a potential novel biomarker for this disease. The presence of a functional pyknon within N-BLR and the related finding that many more pyknon-containing genomic loci in the human genome exhibit tissue-specific and disease-specific expression suggests the possibility of an alternative class of biomarkers and therapeutic targets that are primate-specific. Electronic supplementary material The online version of this article (doi:10.1186/s13059-017-1224-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Isidore Rigoutsos
- Computational Medicine Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA.
| | - Sang Kil Lee
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Present address: Institute of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Su Youn Nam
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Gastroenterology, Department of Internal Medicine, Kyungpook National University Medical School, Daegu, Korea
| | - Simone Anfossi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Barbara Pasculli
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Present address: Laboratory of Oncology, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Martin Pichler
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Present address: Division of Oncology, Medical University of Graz, Graz, Austria
| | - Yi Jing
- Computational Medicine Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Center for RNA interference and non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aristeidis G Telonis
- Computational Medicine Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Simona Rossi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Present address: Institute of Oncology Research (IOR), Research Division of the Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Center for RNA interference and non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tina Catela Ivkovic
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Linda Fabris
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Peter M Clark
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hui Ling
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Masayoshi Shimizu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roxana S Redis
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Present address: ProQR Therapeutics, Leiden, Netherlands
| | - Maitri Y Shah
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xinna Zhang
- Center for RNA interference and non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yoshinaga Okugawa
- Center for Gastrointestinal Research, and Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
| | - Eun Jung Jung
- Department of Surgery, School of Medicine, Gyeongsang National University, Jin-ju, South Korea
| | | | - Li Huang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jana Ferdin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Present address: Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Roberta Gafà
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Riccardo Spizzo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Present address: CRO, National Cancer Institute, 33081, Aviano, Italy
| | - Milena S Nicoloso
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Present address: CRO, National Cancer Institute, 33081, Aviano, Italy
| | - Anurag N Paranjape
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Present address: National Cancer Institute, Bethesda, MD, USA
| | - Maryam Shariati
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aida Tiron
- Department of Medicine, Nassau University Medical Center, 2201 Hempstead Tpke, East Meadow, NY, 11554, USA
| | - Jen Jen Yeh
- Departments of Surgery and Pharmacology, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Raul Teruel-Montoya
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Present address: Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBEER (CB15/00055), Murcia, Spain
| | - Lianchun Xiao
- Division of Quantitative Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sonia A Melo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, and Ipatimup - Institute of Pathology and Molecular Immunology of the University of Porto, 4200, Porto, Portugal.,Department of Pathology, Faculty of Medicine of Porto University, 4200-319, Porto, Portugal
| | - David Menter
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhi-Qin Jiang
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elsa R Flores
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Massimo Negrini
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Ajay Goel
- Center for Gastrointestinal Research, and Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
| | - Menashe Bar-Eli
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chang Gong Liu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Center for RNA interference and non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Medfuture, Cluj-Napoca, Romania.,Research Center for Advanced Medicine - University of Medicine and Pharmacy "Iuliu Haţieganu", Cluj-Napoca, Romania.,Department of Functional Genomics, Proteomics and Experimental Pathology- The Oncology Institute " Prof Dr. Ion Chiricuta, Cluj-Napoca, Romania
| | - Manel Esteller
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Catalonia, Spain.,Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain.,Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giovanni Lanza
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Center for RNA interference and non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
29
|
Rogers ZN, McFarland CD, Winters IP, Naranjo S, Chuang CH, Petrov D, Winslow MM. A quantitative and multiplexed approach to uncover the fitness landscape of tumor suppression in vivo. Nat Methods 2017; 14:737-742. [PMID: 28530655 PMCID: PMC5495136 DOI: 10.1038/nmeth.4297] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 04/19/2017] [Indexed: 12/29/2022]
Abstract
Cancer growth is a multi-stage, stochastic evolutionary process. While cancer genome sequencing has been instrumental in identifying the genomic alterations that occur in human tumors, the consequences of these alterations on tumor growth remains largely unexplored. Conventional genetically engineered mouse models enable the study of tumor growth in vivo, but they are neither readily scalable nor sufficiently quantitative to unravel the magnitude and mode of action of many tumor suppressor genes. Here, we present a method that integrates tumor barcoding with ultra-deep barcode sequencing (Tuba-seq) to interrogate tumor suppressor function in mouse models of human cancer. Tuba-seq uncovers genotype-dependent distributions of tumor sizes with great precision. By combining Tuba-seq with multiplexed CRISPR/Cas9-mediated genome editing, we quantified the effects of eleven tumor-suppressor pathways that are frequently altered in human lung adenocarcinoma. With unprecedented resolution, parallelization, and precision Tuba-seq enables broad quantification of tumor suppressor gene function.
Collapse
Affiliation(s)
- Zoë N Rogers
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | | | - Ian P Winters
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Santiago Naranjo
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Chen-Hua Chuang
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Dmitri Petrov
- Department of Biology, Stanford University, Stanford, California, USA
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, California, USA.,Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
30
|
Application of CRISPR-mediated genome engineering in cancer research. Cancer Lett 2017; 387:10-17. [DOI: 10.1016/j.canlet.2016.03.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 03/12/2016] [Accepted: 03/14/2016] [Indexed: 12/21/2022]
|
31
|
Bizzarri M, Cucina A. SMT and TOFT: Why and How They are Opposite and Incompatible Paradigms. Acta Biotheor 2016; 64:221-39. [PMID: 27283400 DOI: 10.1007/s10441-016-9281-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 05/23/2016] [Indexed: 01/02/2023]
Abstract
The Somatic Mutation Theory (SMT) has been challenged on its fundamentals by the Tissue Organization Field Theory of Carcinogenesis (TOFT). However, a recent publication has questioned whether TOFT could be a valid alternative theory of carcinogenesis to that presented by SMT. Herein we critically review arguments supporting the irreducible opposition between the two theoretical approaches by highlighting differences regarding the philosophical, methodological and experimental approaches on which they respectively rely. We conclude that SMT has not explained carcinogenesis due to severe epistemological and empirical shortcomings, while TOFT is gaining momentum. The main issue is actually to submit SMT to rigorous testing. This concern includes the imperatives to seek evidence for disproving one's hypothesis, and to consider the whole, and not just selective evidence.
Collapse
Affiliation(s)
- Mariano Bizzarri
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
- Systems Biology Group Lab, Sapienza University of Rome, Via Antonio Scarpa 14, 00161, Rome, Italy.
| | - Alessandra Cucina
- Department of Surgery "Pietro Valdoni", Sapienza University of Rome, Via A. Scarpa 14, 00161, Rome, Italy
- Azienda Policlinico Umberto I, Viale del Policlinico 155, 00161, Rome, Italy
| |
Collapse
|
32
|
Li J, Sordella R, Powers S. Effectors and potential targets selectively upregulated in human KRAS-mutant lung adenocarcinomas. Sci Rep 2016; 6:27891. [PMID: 27301828 PMCID: PMC4908391 DOI: 10.1038/srep27891] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/26/2016] [Indexed: 12/11/2022] Open
Abstract
Genetic and proteomic analysis of human tumor samples can provide an important compliment to information obtained from model systems. Here we examined protein and gene expression from the Cancer Genome and Proteome Atlases (TCGA and TCPA) to characterize proteins and protein-coding genes that are selectively upregulated in KRAS-mutant lung adenocarcinomas. Phosphoprotein activation of several MAPK signaling components was considerably stronger in KRAS-mutants than any other group of tumors, even those with activating mutations in receptor tyrosine kinases (RTKs) and BRAF. Co-occurring mutations in KRAS-mutants were associated with differential activation of PDK1 and PKC-alpha. Genes showing strong activation in RNA-seq data included negative regulators of RTK/RAF/MAPK signaling along with potential oncogenic effectors including activators of Rac and Rho proteins and the receptor protein-tyrosine phosphatase genes PTPRM and PTPRE. These results corroborate RAF/MAPK signaling as an important therapeutic target in KRAS-mutant lung adenocarcinomas and pinpoint new potential targets.
Collapse
Affiliation(s)
- Jinyu Li
- Department of Pathology, Stony Brook University, Stony Brook, NY, 11794, USA
| | | | - Scott Powers
- Department of Pathology, Stony Brook University, Stony Brook, NY, 11794, USA.,Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| |
Collapse
|
33
|
Pore N, Jalla S, Liu Z, Higgs B, Sorio C, Scarpa A, Hollingsworth R, Tice DA, Michelotti E. In Vivo Loss of Function Screening Reveals Carbonic Anhydrase IX as a Key Modulator of Tumor Initiating Potential in Primary Pancreatic Tumors. Neoplasia 2016; 17:473-80. [PMID: 26152355 PMCID: PMC4719001 DOI: 10.1016/j.neo.2015.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/02/2015] [Accepted: 05/05/2015] [Indexed: 02/06/2023] Open
Abstract
Reprogramming of energy metabolism is one of the emerging hallmarks of cancer. Up-regulation of energy metabolism pathways fuels cell growth and division, a key characteristic of neoplastic disease, and can lead to dependency on specific metabolic pathways. Thus, targeting energy metabolism pathways might offer the opportunity for novel therapeutics. Here, we describe the application of a novel in vivo screening approach for the identification of genes involved in cancer metabolism using a patient-derived pancreatic xenograft model. Lentiviruses expressing short hairpin RNAs (shRNAs) targeting 12 different cell surface protein transporters were separately transduced into the primary pancreatic tumor cells. Transduced cells were pooled and implanted into mice. Tumors were harvested at different times, and the frequency of each shRNA was determined as a measure of which ones prevented tumor growth. Several targets including carbonic anhydrase IX (CAIX), monocarboxylate transporter 4, and anionic amino acid transporter light chain, xc- system (xCT) were identified in these studies and shown to be required for tumor initiation and growth. Interestingly, CAIX was overexpressed in the tumor initiating cell population. CAIX expression alone correlated with a highly tumorigenic subpopulation of cells. Furthermore, CAIX expression was essential for tumor initiation because shRNA knockdown eliminated the ability of cells to grow in vivo. To the best of our knowledge, this is the first parallel in vivo assessment of multiple novel oncology target genes using a patient-derived pancreatic tumor model.
Collapse
Affiliation(s)
| | | | - Zheng Liu
- MedImmune, LLC, Gaithersburg, MD, USA
| | | | - Claudio Sorio
- ARC-NET Research Centre and Department of Pathology and Diagnostics, University of Verona Medical School, Verona, Italy
| | - Aldo Scarpa
- ARC-NET Research Centre and Department of Pathology and Diagnostics, University of Verona Medical School, Verona, Italy
| | | | | | | |
Collapse
|
34
|
Abstract
How can we stop cancer progression? Current strategies depend on modelling progression as the balanced outcome of mutations in, and expression of, tumour suppressor genes and oncogenes. New treatments emerge from successful attempts to tip that balance, but secondary mutational escape from those treatments has become a major impediment because it leads to resistance. In this Opinion article, we argue for a return to an earlier stratagem: tumour cell reversion. Treatments based on selection and analysis of stable revertants could create more durable remissions by reducing the selective pressure that leads to rapid drug resistance.
Collapse
|
35
|
Abstract
As cancer has become increasingly prevalent, cancer prevention research has evolved towards placing a greater emphasis on reducing cancer deaths and minimizing the adverse consequences of having cancer. 'Precision cancer prevention' takes into account the collaboration of intrinsic and extrinsic factors in influencing cancer incidence and aggressiveness in the context of the individual, as well as recognizing that such knowledge can improve early detection and enable more accurate discrimination of cancerous lesions. However, mouse models, and particularly genetically engineered mouse (GEM) models, have yet to be fully integrated into prevention research. In this Opinion article, we discuss opportunities and challenges for precision mouse modelling, including the essential criteria of mouse models for prevention research, representative success stories and opportunities for more refined analyses in future studies.
Collapse
Affiliation(s)
| | - Aditya Dutta
- Department of Urology, Columbia University Medical Center, New York, NY 10032
| | - Cory Abate-Shen
- Department of Urology, Columbia University Medical Center, New York, NY 10032
- Department of Medicine, Columbia University Medical Center, New York, NY 10032
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY 10032
- Department of Institute of Cancer Genetics, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
- Corresponding author: Cory Abate-Shen, Columbia University Medical Center, 1130 St. Nicholas Ave., New York, NY 10032, (CAS) Phone: (212) 851-4731; fax: (212) 851-4787;
| |
Collapse
|
36
|
Sanmamed MF, Chester C, Melero I, Kohrt H. Defining the optimal murine models to investigate immune checkpoint blockers and their combination with other immunotherapies. Ann Oncol 2016; 27:1190-8. [PMID: 26912558 DOI: 10.1093/annonc/mdw041] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 01/22/2016] [Indexed: 12/31/2022] Open
Abstract
The recent success of checkpoint blockers to treat cancer has demonstrated that the immune system is a critical player in the war against cancer. Historically, anticancer therapeutics have been tested in syngeneic mouse models (with a fully murine immune system) or in immunodeficient mice that allow the engraftment of human xenografts. Animal models with functioning human immune systems are critically needed to more accurately recapitulate the complexity of the human tumor microenvironment. Such models are integral to better predict tumor responses to both immunomodulatory agents and directly antineoplastic therapies. In this regard, the development of humanized models is a promising, novel strategy that offers the possibility of testing checkpoint blockers' capacity and their combination with other antitumor drugs. In this review, we discuss the strengths and weaknesses of the available animal models regarding their capacity to evaluate checkpoint blockers and checkpoint blocker-based combination immunotherapy.
Collapse
Affiliation(s)
- M F Sanmamed
- Department of Immunobiology, Yale University School of Medicine, New Haven
| | - C Chester
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, USA
| | - I Melero
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - H Kohrt
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, USA
| |
Collapse
|
37
|
Animal models of colorectal cancer with liver metastasis. Cancer Lett 2016; 387:114-120. [PMID: 26850374 DOI: 10.1016/j.canlet.2016.01.048] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/26/2016] [Accepted: 01/26/2016] [Indexed: 01/01/2023]
Abstract
Liver metastasis is a leading cause of death in patients with colorectal cancer. Investigating the mechanisms of liver metastasis and control of disease progression are important strategies for improving survival of these patients. Liver metastasis is a multi-step process and relevant models representing these steps are necessary to understand the mechanism of liver metastasis and establish appropriate treatments. Recently, the development of animal models for use in metastasis research has greatly increased; however, there is still a lack of models that sufficiently represent human cancer. Thus, in order to select an optimal model for of a given study, it is necessary to fully understand the characteristics of each animal model. In this review, we describe the mouse models currently used for colorectal cancer with liver metastasis, their characteristics, and their pros and cons. This may help us specify the mechanism of liver metastasis and provide evidence relevant to clinical applications.
Collapse
|
38
|
Day CP, Merlino G, Van Dyke T. Preclinical mouse cancer models: a maze of opportunities and challenges. Cell 2015; 163:39-53. [PMID: 26406370 DOI: 10.1016/j.cell.2015.08.068] [Citation(s) in RCA: 402] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Indexed: 12/20/2022]
Abstract
Significant advances have been made in developing novel therapeutics for cancer treatment, and targeted therapies have revolutionized the treatment of some cancers. Despite the promise, only about five percent of new cancer drugs are approved, and most fail due to lack of efficacy. The indication is that current preclinical methods are limited in predicting successful outcomes. Such failure exacts enormous cost, both financial and in the quality of human life. This Primer explores the current status, promise, and challenges of preclinical evaluation in advanced mouse cancer models and briefly addresses emerging models for early-stage preclinical development.
Collapse
Affiliation(s)
- Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| | - Terry Van Dyke
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
39
|
Peela N, Sam FS, Christenson W, Truong D, Watson AW, Mouneimne G, Ros R, Nikkhah M. A three dimensional micropatterned tumor model for breast cancer cell migration studies. Biomaterials 2015; 81:72-83. [PMID: 26724455 DOI: 10.1016/j.biomaterials.2015.11.039] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 11/29/2015] [Indexed: 12/14/2022]
Abstract
Breast cancer cell invasion is a highly orchestrated process driven by a myriad of complex microenvironmental stimuli, making it difficult to isolate and assess the effects of biochemical or biophysical cues (i.e. tumor architecture, matrix stiffness) on disease progression. In this regard, physiologically relevant tumor models are becoming instrumental to perform studies of cancer cell invasion within well-controlled conditions. Herein, we explored the use of photocrosslinkable hydrogels and a novel, two-step photolithography technique to microengineer a 3D breast tumor model. The microfabrication process enabled precise localization of cell-encapsulated circular constructs adjacent to a low stiffness matrix. To validate the model, breast cancer cell lines (MDA-MB-231, MCF7) and non-tumorigenic mammary epithelial cells (MCF10A) were embedded separately within the tumor model, all of which maintained high viability throughout the experiments. MDA-MB-231 cells exhibited extensive migratory behavior and invaded the surrounding matrix, whereas MCF7 or MCF10A cells formed clusters that stayed confined within the circular tumor regions. Additionally, real-time cell tracking indicated that the speed and persistence of MDA-MB-231 cells were substantially higher within the surrounding matrix compared to the circular constructs. Z-stack imaging of F-actin/α-tubulin cytoskeletal organization revealed unique 3D protrusions in MDA-MB-231 cells and an abundance of 3D clusters formed by MCF7 and MCF10A cells. Our results indicate that gelatin methacrylate (GelMA) hydrogel, integrated with the two-step photolithography technique, has great promise in the development of 3D tumor models with well-defined architecture and tunable stiffness.
Collapse
Affiliation(s)
- Nitish Peela
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ 85287, USA
| | - Feba S Sam
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ 85287, USA
| | - Wayne Christenson
- Center for Biological Physics, Arizona State University, Tempe, AZ 85287, USA; Department of Physics, Arizona State University, Tempe, AZ 85287, USA
| | - Danh Truong
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ 85287, USA
| | - Adam W Watson
- University of Arizona Cancer Center, Department of Cellular and Molecular Medicine, Tucson, AZ 85724, USA
| | - Ghassan Mouneimne
- University of Arizona Cancer Center, Department of Cellular and Molecular Medicine, Tucson, AZ 85724, USA
| | - Robert Ros
- Center for Biological Physics, Arizona State University, Tempe, AZ 85287, USA; Department of Physics, Arizona State University, Tempe, AZ 85287, USA; Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ 85287, USA.
| |
Collapse
|
40
|
Hou X, Du Y, Deng Y, Wu J, Cao G. Sleeping Beauty transposon system for genetic etiological research and gene therapy of cancers. Cancer Biol Ther 2015; 16:8-16. [PMID: 25455252 DOI: 10.4161/15384047.2014.986944] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Carcinogenesis is etiologically associated with somatic mutations of critical genes. Recently, a number of somatic mutations and key molecules have been found to be involved in functional networks affecting cancer progression. Suitable animal models are required to validate cancer-promoting or -inhibiting capacities of these mutants and molecules. Sleeping Beauty transposon system consists of a transposon that carries gene(s) of interest and a transposase that recognizes, excises, and reinserts genes in given location of the genome. It can create both gain-of-function and loss-of-function mutations, thus being frequently chosen to investigate the etiological mechanisms and gene therapy for cancers in animal models. In this review, we summarized current advances of Sleeping Beauty transposon system in revealing molecular mechanism of cancers and improving gene therapy. Understanding molecular mechanisms by which driver mutations contribute to carcinogenesis and metastasis may pave the way for the development of innovative prophylactic and therapeutic strategies against malignant diseases.
Collapse
Key Words
- 7, 12-dimethylbenzanthracene/12-O-tetradecanoylphorbol-13-acetate
- Alb-Cre, Albumin promoter-Cre
- CAG promoter, CMV enhancer/chicken β-actin promoter
- CAR, chimeric antigen receptor
- CIS, common insertion site
- CMV, chimeric cytomegalovirus
- CRC, colorectal cancer
- Cre, cyclization recombination enzyme
- DDE, Asp, Asp, Glu
- DMBA/TPA
- DR, direct orientation
- Fah, fumarylacetoacetate hydrolase gene
- GWAS, gnome wide analysis study
- HBV, Hepatitis B Virus
- HBx, HBV X protein
- HCC, hepatocellular carcinoma
- IRs, inverted repeat sequences
- LsL, loxP-stop-loxP
- MPNSTs, malignant peripheral nerve sheath tumor
- MSCV, murine stem cell virus
- PAI, Pro, Ala, Ile
- PBMCs, peripheral blood mononuclear cells
- RED, Arg, Glu, Asp
- RosaSBaseLsL, Cre-inducible SBase allele
- Rtl1, Retrotransposon-like 1
- SB, Sleeping Beauty
- SBase, Sleeping Beauty transposase
- Sleeping Beauty transposon system
- StatinAE, angiostatin-endostatin fusion gene
- Trp53, transformation related protein 53
- animal model
- driver
- gene function
- gene therapy
- malignant diseases
- sgRNA, single guide RNA
- shp53, short hairpin RNA against the Trp53 gene
- somatic mutation
Collapse
Affiliation(s)
- Xiaomei Hou
- a Department of Epidemiology ; Second Military Medical University ; Shanghai , China
| | | | | | | | | |
Collapse
|
41
|
Kim SB, Bozeman RG, Kaisani A, Kim W, Zhang L, Richardson JA, Wright WE, Shay JW. Radiation promotes colorectal cancer initiation and progression by inducing senescence-associated inflammatory responses. Oncogene 2015; 35:3365-75. [PMID: 26477319 PMCID: PMC4837107 DOI: 10.1038/onc.2015.395] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 08/27/2015] [Accepted: 09/08/2015] [Indexed: 12/29/2022]
Abstract
Proton radiotherapy is becoming more common since protons induce more precise DNA damage at the tumor site with reduced side effects to adjacent normal tissues. However, the long-term biological effects of proton irradiation in cancer initiation compared to conventional photon irradiation are poorly characterized. In this study, using a human familial adenomatous polyposis syndrome susceptible mouse model, we show that whole body irradiation with protons are more effective in inducing senescence-associated inflammatory responses (SIR) which are involved in colon cancer initiation and progression. After proton irradiation, a subset of SIR genes (Troy, Sox17, Opg, Faim2, Lpo, Tlr2 and Ptges) and a gene known to be involved in invasiveness (Plat), along with the senescence associated gene (P19Arf) are markedly increased. Following these changes loss of Casein kinase Iα (CKIα) and induction of chronic DNA damage and TP53 mutations are increased compared to x-ray irradiation. Proton irradiation also increases the number of colonic polyps, carcinomas and invasive adenocarcinomas. Pretreatment with the non-steroidal anti-inflammatory drug, CDDO-EA, reduces proton irradiation associated SIR and tumorigenesis. Thus, exposure to proton irradiation elicits significant changes in colorectal cancer initiation and progression that can be mitigated using CDDO-EA.
Collapse
Affiliation(s)
- S B Kim
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - R G Bozeman
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - A Kaisani
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - W Kim
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - L Zhang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - J A Richardson
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - W E Wright
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - J W Shay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| |
Collapse
|
42
|
Kemp CJ. Animal Models of Chemical Carcinogenesis: Driving Breakthroughs in Cancer Research for 100 Years. Cold Spring Harb Protoc 2015; 2015:865-74. [PMID: 26430259 PMCID: PMC4949043 DOI: 10.1101/pdb.top069906] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The identification of carcinogens in the workplace, diet, and environment through chemical carcinogenesis studies in animals has directly contributed to a reduction of cancer burden in the human population. Reduced exposure to these carcinogens through lifestyle changes, government regulation, or change in industry practices has reduced cancer incidence in exposed populations. In addition to providing the first experimental evidence for cancer's relationship to chemical and radiation exposure, animal models of environmentally induced cancer have and will continue to provide important insight into the causes, mechanisms, and conceptual frameworks of cancer. More recently, combining chemical carcinogens with genetically engineered mouse models has emerged as an invaluable approach to study the complex interaction between genotype and environment that contributes to cancer development. In the future, animal models of environmentally induced cancer are likely to provide insight into areas such as the epigenetic basis of cancer, genetic modifiers of cancer susceptibility, the systems biology of cancer, inflammation and cancer, and cancer prevention.
Collapse
Affiliation(s)
- Christopher J Kemp
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| |
Collapse
|
43
|
Liu C, Li Z, Zhang Y. [Application Progress of CRISPR/Cas9 System for Gene Editing in Tumor Research]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2015; 18:571-9. [PMID: 26383982 PMCID: PMC6000117 DOI: 10.3779/j.issn.1009-3419.2015.09.08] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
CRISPR/Cas9(Clustered Regularly Interspaced Short Palindromic Repeat/CRISPR-associated nuclease 9)基因编辑系统是基于古细菌抵御外源核酸入侵的免疫机制为基础开发出来的一种新型的基因编辑技术。相对于传统的基因编辑系统,该系统具有更加高效、操作简单、细胞毒性小等特点。目前,CRISPR/Cas9基因编辑技术已经在肿瘤研究的诸多方面中得到应用,包括肿瘤相关基因的功能研究、构建动物肿瘤模型、筛选肿瘤细胞表型及耐药相关基因以及肿瘤的基因治疗等诸多方面。本文就其在肿瘤研究中的应用进展进行综述。
Collapse
Affiliation(s)
- Chao Liu
- Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Zhiwei Li
- Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Yanqiao Zhang
- Harbin Medical University Cancer Hospital, Harbin 150081, China
| |
Collapse
|
44
|
Yan Y, Chen N, Wang Y, Wang K. The application of antitumor drug-targeting models on liver cancer. Drug Deliv 2015; 23:1667-75. [PMID: 26289213 DOI: 10.3109/10717544.2015.1064188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Hepatocarcinoma animal models, such as the induced tumor model, transplanted tumor model, gene animal model, are significant experimental tools for the evaluation of targeting drug delivery system as well as the pre-clinical studies of liver cancer. The application of antitumor drug-targeting models not only furnishes similar biological characteristics to human liver cancer but also offers guarantee of pharmacokinetic indicators of the liver-targeting preparations. In this article, we have reviewed some kinds of antitumor drug-targeting models of hepatoma and speculated that the research on this field would be capable of attaining a deeper level and expecting a superior achievement in the future.
Collapse
Affiliation(s)
- Yan Yan
- a School of Pharmacy, Xi'an Jiaotong University , Xi'an , Shaanxi , China
| | - Ningbo Chen
- b Surgical Department of Emergency Center, The People's Hospital of Sichuan Province , Sichuan , China and
| | - Yunbing Wang
- c National Engineering Research Center in Biomaterials, Sichuan University , Chengdu , Sichuan , China
| | - Ke Wang
- a School of Pharmacy, Xi'an Jiaotong University , Xi'an , Shaanxi , China
| |
Collapse
|
45
|
Mou H, Kennedy Z, Anderson DG, Yin H, Xue W. Precision cancer mouse models through genome editing with CRISPR-Cas9. Genome Med 2015; 7:53. [PMID: 26060510 PMCID: PMC4460969 DOI: 10.1186/s13073-015-0178-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The cancer genome is highly complex, with hundreds of point mutations, translocations, and chromosome gains and losses per tumor. To understand the effects of these alterations, precise models are needed. Traditional approaches to the construction of mouse models are time-consuming and laborious, requiring manipulation of embryonic stem cells and multiple steps. The recent development of the clustered regularly interspersed short palindromic repeats (CRISPR)-Cas9 system, a powerful genome-editing tool for efficient and precise genome engineering in cultured mammalian cells and animals, is transforming mouse-model generation. Here, we review how CRISPR-Cas9 has been used to create germline and somatic mouse models with point mutations, deletions and complex chromosomal rearrangements. We highlight the progress and challenges of such approaches, and how these models can be used to understand the evolution and progression of individual tumors and identify new strategies for cancer treatment. The generation of precision cancer mouse models through genome editing will provide a rapid avenue for functional cancer genomics and pave the way for precision cancer medicine.
Collapse
Affiliation(s)
- Haiwei Mou
- RNA Therapeutics Institute and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605 USA
| | - Zachary Kennedy
- RNA Therapeutics Institute and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605 USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142 USA ; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142 USA ; Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA 02139 USA ; Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142 USA
| | - Hao Yin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142 USA
| | - Wen Xue
- RNA Therapeutics Institute and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605 USA
| |
Collapse
|
46
|
Fernandes E, Ferreira JA, Andreia P, Luís L, Barroso S, Sarmento B, Santos LL. New trends in guided nanotherapies for digestive cancers: A systematic review. J Control Release 2015; 209:288-307. [PMID: 25957905 DOI: 10.1016/j.jconrel.2015.05.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 05/02/2015] [Accepted: 05/05/2015] [Indexed: 02/06/2023]
Abstract
Digestive tract tumors are among the most common and deadliest malignancies worldwide, mainly due to late diagnosis and lack of efficient therapeutics. Current treatments essentially rely on surgery associated with (neo)adjuvant chemotherapy agents. Despite an upfront response, conventional drugs often fail to eliminate highly aggressive clones endowed with chemoresistant properties, which are responsible for tumor recurrence and disease dissemination. Synthetic drugs also present severe adverse systemic effects, hampering the administration of biologically effective dosages. Nanoencapsulation of chemotherapeutic agents within biocompatible polymeric or lipid matrices holds great potential to improve the pharmacokinetics and efficacy of conventional chemotherapy while reducing systemic toxicity. Tagging nanoparticle surfaces with specific ligands for cancer cells, namely monoclonal antibodies or antibody fragments, has provided means to target more aggressive clones, further improving the selectivity and efficacy of nanodelivery vehicles. In fact, over the past twenty years, significant research has translated into a wide array of guided nanoparticles, providing the molecular background for a new generation of intelligent and more effective anti-cancer agents. Attempting to bring awareness among the medical community to emerging targeted nanopharmaceuticals and foster advances in the field, we have conducted a systematic review about this matter. Emphasis was set on ongoing preclinical and clinical trials for liver, colorectal, gastric and pancreatic cancers. To the best of our knowledge this is the first systematic and integrated overview on this field. Using a specific query, 433 abstracts were gathered and narrowed to 47 manuscripts when matched against inclusion/exclusion criteria. All studies showed that active targeting improves the effectiveness of the nanodrugs alone, while lowering its side effects. The main focus has been on hepatocarcinomas, mainly by exploring glycans as homing molecules. Other ligands such as peptides/small proteins and antibodies/antibody fragments, with affinity to either tumor vasculature or tumor cells, have also been widely and successfully applied to guide nanodrugs to gastrointestinal carcinomas. Conversely, few solutions have been presented for pancreatic tumors. To this date only three nanocomplexes have progressed beyond pre-clinical stages: i) PK2, a galactosamine-functionalized polymeric-DOX formulation for hepatocarcinomas; ii) MCC-465, an anti-(myosin heavy chain a) immunoliposome for advanced stage metastatic solid tumors; and iii) MBP-426, a transferrin-liposome-oxaliplatin conjugate, also for advanced stage tumors. Still, none has been approved for clinical use. However, based on the high amount of pre-clinical studies showing enthusiastic results, the number of clinical trials is expected to increase in the near future. A more profound understanding about the molecular nature of chemoresistant clones and cancer stem cell biology will also contribute to boost the field of guided nanopharmacology towards more effective solutions.
Collapse
Affiliation(s)
- Elisabete Fernandes
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal; I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal and INEB - Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal; Mass Spectrometry Center, QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal.
| | - Peixoto Andreia
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal
| | - Lima Luís
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal; Nucleo de Investigação em Farmácia - Centro de Investigação em Saúde e Ambiente (CISA), Health School of the Polytechnic Institute of Porto, Porto, Portugal
| | - Sérgio Barroso
- Serviço de Oncologia, Hospital de Évora, Évora, Portugal
| | - Bruno Sarmento
- I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal and INEB - Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal; CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Gandra PRD, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal; Health School of University of Fernando Pessoa, Porto, Portugal; Department of Surgical Oncology, Portuguese Institute of Oncology, Porto, Portugal
| |
Collapse
|
47
|
Barck KH, Bou-Reslan H, Rastogi U, Sakhuja T, Long JE, Molina R, Lima A, Hamilton P, Junttila MR, Johnson L, Carano RAD. Quantification of Tumor Burden in a Genetically Engineered Mouse Model of Lung Cancer by Micro-CT and Automated Analysis. Transl Oncol 2015; 8:126-35. [PMID: 25926079 PMCID: PMC4415142 DOI: 10.1016/j.tranon.2015.03.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/06/2015] [Accepted: 03/17/2015] [Indexed: 11/18/2022] Open
Abstract
Genetically engineered mouse models (GEMMs) of lung cancer closely recapitulate the human disease but suffer from the difficulty of evaluating tumor growth by conventional methods. Herein, a novel automated image analysis method for estimating the lung tumor burden from in vivo micro-computed tomography (micro-CT) data is described. The proposed tumor burden metric is the segmented soft tissue volume contained within a chest space region of interest, excluding an estimate of the heart volume. The method was validated by comparison with previously published manual analysis methods and applied in two therapeutic studies in a mutant K-ras GEMM of non-small cell lung carcinoma. Mice were imaged by micro-CT pre-treatment and stratified into four treatment groups: an antibody inhibiting vascular endothelial growth factor (anti-VEGF), chemotherapy, combination of anti-VEGF and chemotherapy, or control antibody. In the first study, post-treatment imaging was performed 4 weeks later. In the second study, mice were scanned serially on a high-throughput scanner every 2 weeks for 8 weeks during treatment. In both studies, the automated tumor burden estimates were well correlated with manual metrics (r value range: 0.83-0.93, P < .0001) and showed a similar, significant reduction in tumor growth in mice treated with anti-VEGF alone or in combination with chemotherapy. Given the fully automated nature of this technique, the proposed analysis method can provide a valuable tool in preclinical drug research for screening and randomizing animals into treatment groups and evaluating treatment efficacy in mouse models of lung cancer in a highly robust and efficient manner.
Collapse
Affiliation(s)
- Kai H Barck
- Department of Biomedical Imaging, Genentech, Inc, South San Francisco, CA, USA
| | - Hani Bou-Reslan
- Department of Biomedical Imaging, Genentech, Inc, South San Francisco, CA, USA
| | - Ujjawal Rastogi
- Department of Biomedical Imaging, Genentech, Inc, South San Francisco, CA, USA
| | - Timothy Sakhuja
- Department of Biomedical Imaging, Genentech, Inc, South San Francisco, CA, USA
| | - Jason E Long
- Department of Translational Oncology, Genentech, Inc, South San Francisco, CA, USA
| | - Rafael Molina
- Department of Translational Oncology, Genentech, Inc, South San Francisco, CA, USA
| | - Anthony Lima
- Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA, USA
| | - Patricia Hamilton
- Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA, USA
| | - Melissa R Junttila
- Department of Translational Oncology, Genentech, Inc, South San Francisco, CA, USA
| | - Leisa Johnson
- Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA, USA
| | - Richard A D Carano
- Department of Biomedical Imaging, Genentech, Inc, South San Francisco, CA, USA.
| |
Collapse
|
48
|
Wang Y, Tseng JC, Sun Y, Beck AH, Kung AL. Noninvasive imaging of tumor burden and molecular pathways in mouse models of cancer. Cold Spring Harb Protoc 2015; 2015:135-44. [PMID: 25646505 DOI: 10.1101/pdb.top069930] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Imaging plays a central role in the diagnosis of cancer and the evaluation of therapeutic efficacy in patients with cancer. Because macroscopic imaging is noninvasive and quantitative, the development of specialized instruments for small animals has spurred increasing utilization in preclinical cancer studies. Some small-animal imaging devices are miniaturized derivatives of clinical imaging modalities, including computed tomography, magnetic resonance imaging, positron-emission tomography, single-photon emission computed tomography, and ultrasonography. Optical imaging, including bioluminescence imaging and fluorescence imaging, has evolved from microscopic cellular imaging technologies. Here, we review how current imaging modalities are enabling high-resolution structural imaging with micrometer-scale spatial resolution, thus allowing for the quantification of tumor burden in genetically engineered and orthotopic models of cancer, where tumors develop within organs not typically accessible to measurements with calipers. Beyond measuring tumor size, imaging is increasingly being used to assess the activity of molecular pathways within tumors and to reveal the pharmacodynamic efficacy of targeted therapies. Each imaging technology has particular strengths and limitations, and we discuss how studies should be carefully designed to match the imaging approach to the primary experimental question.
Collapse
Affiliation(s)
- Yuchuan Wang
- Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts 02215
| | - Jen-Chieh Tseng
- Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts 02215
| | - Yanping Sun
- Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts 02215
| | - Andrew H Beck
- Department of Pathology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215
| | - Andrew L Kung
- Department of Pediatrics, Columbia University Medical Center, New York, New York 10032
| |
Collapse
|
49
|
Abstract
Breast cancer is the most common cause of cancer death in women worldwide. This malignancy is a complex disease, which is defined by an intrinsic heterogeneity on the histopathological and molecular level as well as response to therapy and outcome. In addition to classical histopathological features, breast cancer can be categorized into at least five major subtypes based on comprehensive gene expression profiling: luminal A, luminal B, basal-like, ERBB2-positive, and normal-like breast cancer. Genetically engineered mouse models can serve as tools to study the molecular underpinnings for this disease. Given the genetic complexity that drives the initiation and progression of individual breast cancer subtypes, it is evident that certain models can reflect only particular aspects of this malignancy. In this book chapter, we will primarily focus on advances in modeling breast cancer at defined stages of carcinogenesis using genetically engineered mice. We will discuss the ability as well as shortcomings of these models to faithfully recapitulate the spectrum of human breast cancer subtypes.
Collapse
|
50
|
Gad E, Rastetter L, Slota M, Koehnlein M, Treuting PM, Dang Y, Stanton S, Disis ML. Natural history of tumor growth and immune modulation in common spontaneous murine mammary tumor models. Breast Cancer Res Treat 2014; 148:501-10. [PMID: 25395320 PMCID: PMC4280094 DOI: 10.1007/s10549-014-3199-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 11/04/2014] [Indexed: 12/20/2022]
Abstract
Recent studies in patients with breast cancer suggest the immune microenvironment influences response to therapy. We aimed to evaluate the relationship between growth rates of tumors in common spontaneous mammary tumor models and immune biomarkers evaluated in the tumor and blood. TgMMTV-neu and C3(1)-Tag transgenic mice were followed longitudinally from birth, and MPA-DMBA-treated mice from the time of carcinogen administration, for the development of mammary tumors. Tumor-infiltrating CD4(+) and CD8(+) T-cells, FOXP3(+) T-regulatory cells, and myeloid-derived suppressor cells were assessed by flow cytometry. Serum cytokines were evaluated in subsets of mice. Fine needle aspirates of tumors were collected and RNA was isolated to determine levels of immune and proliferation markers. Age of tumor onset and kinetics of tumor growth were significantly different among the models. Mammary tumors from TgMMTV-neu contained a lower CD8/CD4 ratio than that of other models (p < 0.05). MPA-DMBA-induced tumors contained a higher percentage of FOXP3(+) CD4(+) T-cells (p < 0.01) and MDSC (p < 0.001) compared with the other models. Individuals with significantly slower tumor growth demonstrated higher levels of Type I serum cytokines prior to the development of lesions compared to those with rapid tumor growth. Moreover, the tumors of animals with more rapid tumor growth demonstrated a significant increase in the expression of genes associated with Type II immunity than those with slower-progressing tumors. These data provide a foundation for the development of in vivo models to explore the relationship between endogenous immunity and response to standard therapies for breast cancer.
Collapse
Affiliation(s)
- Ekram Gad
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, Seattle, WA
| | - Lauren Rastetter
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, Seattle, WA
| | - Meredith Slota
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, Seattle, WA
| | - Marlese Koehnlein
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, Seattle, WA
| | - Piper M. Treuting
- Department of Comparative Medicine, University of Washington, Seattle, WA
| | - Yushe Dang
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, Seattle, WA
| | - Sasha Stanton
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, Seattle, WA
| | - Mary L. Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, Seattle, WA
| |
Collapse
|