101
|
Li S, Zhang Z, Han L. 3D Spheroids Propel Tumor Characterization. Trends Cancer 2020; 6:622-624. [DOI: 10.1016/j.trecan.2020.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/13/2022]
|
102
|
Choi EW, Lee M, Song JW, Kim K, Lee J, Yang J, Lee SH, Kim IY, Choi JH, Seong JK. Fas mutation reduces obesity by increasing IL-4 and IL-10 expression and promoting white adipose tissue browning. Sci Rep 2020; 10:12001. [PMID: 32686763 PMCID: PMC7371740 DOI: 10.1038/s41598-020-68971-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 06/23/2020] [Indexed: 12/15/2022] Open
Abstract
Brown adipose tissue generates heat via the mitochondrial uncoupling protein UCP1 to protect against obesity and hypothermia. Fas mutant MRL/lpr mice exhibit a significantly leaner phenotype compared to wild type MRL/MpJ mice. In this study, we evaluated the inflammatory cell population in the adipose tissue of MRL/lpr mice, which could potentially influence their lean phenotype. Furthermore, we compared beige fat activity between the MRL/MpJ and MRL/lpr mice. Fas mutation resulted in high body temperature, improved glucose tolerance, and decreased fat mass and adipocyte size. Fas mutation prevented high-fat diet-induced obesity and decreased the white adipose tissue M1:M2 ratio. When mice were fed a high-fat diet, UCP1, IL-4, IL-10, and tyrosine hydroxylase genes had significantly higher expression in Fas-mutant mice than in wild type mice. After a cold challenge, UCP1 expression and browning were also significantly higher in the Fas-mutant mice. In summary, Fas-mutant mice are resistant to high-fat diet-induced obesity due to increased IL-4 and IL-10 levels and the promotion of thermogenic protein activity and browning in their adipose tissues. STAT6 activation might contribute to M2 polarisation by increasing IL-4 and IL-10 levels while increases in M2 and tyrosine hydroxylase levels promote browning in response to Fas mutation.
Collapse
Affiliation(s)
- Eun Wha Choi
- Department of Veterinary Clinical Pathology, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, Republic of Korea. .,Laboratory Animal Research Center, Samsung Biomedical Research Institute, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
| | - Minjae Lee
- Laboratory Animal Research Center, Samsung Biomedical Research Institute, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Ji Woo Song
- Laboratory Animal Research Center, Samsung Biomedical Research Institute, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Kyeongdae Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Jungmin Lee
- Laboratory Animal Research Center, Samsung Biomedical Research Institute, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.,Laboratory of Developmental Biology and Genomics, BK21 Plus Program for Advanced Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, and Korea Mouse Phenotyping Center, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Jehoon Yang
- Laboratory Animal Research Center, Samsung Biomedical Research Institute, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Seo Hyun Lee
- Laboratory of Developmental Biology and Genomics, BK21 Plus Program for Advanced Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, and Korea Mouse Phenotyping Center, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Il Yong Kim
- Laboratory of Developmental Biology and Genomics, BK21 Plus Program for Advanced Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, and Korea Mouse Phenotyping Center, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Jae-Hoon Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, BK21 Plus Program for Advanced Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, and Korea Mouse Phenotyping Center, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea. .,Interdiscplinary Program for Bioinformatics, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
103
|
Nam H, Funamoto K, Jeon JS. Cancer cell migration and cancer drug screening in oxygen tension gradient chip. BIOMICROFLUIDICS 2020; 14:044107. [PMID: 32742536 PMCID: PMC7375834 DOI: 10.1063/5.0011216] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/28/2020] [Indexed: 05/07/2023]
Abstract
Cancer metastasis, which is prevalent in malignant tumors, is present in a variety of cases depending on the primary tumor and metastatic site. The cancer metastasis is affected by various factors that surround and constitute a tumor microenvironment. One of the several factors, oxygen tension, can affect cancer cells and induce changes in many ways, including motility, directionality, and viability. In particular, the oxygen tension gradient is formed within a tumor cluster and oxygen is lower toward the center of the cluster from the perivascular area. The simple and efficient designing of the tumor microenvironment using microfluidic devices enables the simplified and robust platform of the complex in vivo microenvironment while observing a clear cause-and-effect between the properties of cancer cells under oxygen tension. Here, a microfluidic device with five channels including a gel channel, media channels, and gas channels is designed. MDA-MB-231cells are seeded in the microfluidic device with hydrogel to simulate their three-dimensional movement in the body. The motility and directionality of the cancer cells under the normoxic and oxygen tension gradient conditions are compared. Also, the viability of the cancer cells is analyzed for each condition when anticancer drugs are applied. Unlike the normoxic condition, under the oxygen tension gradient, cancer cells showed directionality toward higher oxygen tension and decreased viability against the certain anticancer drug. The simplified design of the tumor microenvironment through microfluidic devices enables comprehension of the response of cancer cells to varying oxygen tensions and cancer drugs in the hypoxic tumor microenvironment.
Collapse
Affiliation(s)
- Hyeono Nam
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | | | | |
Collapse
|
104
|
Galectins in the Tumor Microenvironment: Focus on Galectin-1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1259:17-38. [DOI: 10.1007/978-3-030-43093-1_2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
105
|
Kita Y, Saito R, Inoue T, Kim WY, Ogawa O, Kobayashi T. Patient-Derived Urothelial Cancer Xenograft Models: A Systematic Review and Future Perspectives. Bladder Cancer 2020. [DOI: 10.3233/blc-200281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Lack of appropriate models that recapitulate the diversity, heterogeneity, and tumor microenvironment of urothelial cancer (UC) is a limitation to preclinical models. Patient-derived xenograft (PDX) models are a promising tool to overcome some of these issues, and thus we present an up-to-date and comprehensive overview of UC PDX models to aid in their future use. OBJECTIVE: To provide an overview on methodology, applications and limitations as well as future perspectives on bladder cancer PDX models. METHODS: Literature searches using PubMed and Web of Science databases were performed for relevant articles according to the following MeSH terms: “urothelial carcinoma(s)” OR “urothelial cancer” OR “urothelial tumor” OR “bladder cancer(s)” OR “bladder carcinoma(s)” OR “transitional cell carcinoma(s)” AND “xenograft(s)” OR “xenotransplant” at December 6th, 2019. We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. RESULTS: Of the 49 studies extracted, 41 studies after the year 2000 were finally analyzed. Published studies show that (1) UC PDX platforms retained the histology and genomic characteristics of the corresponding patient tumors. (2) UC PDX can be applied to ask various questions including to study the mechanisms of disease progression and treatment resistance, to develop novel drugs and biomarkers, as well as to potentially realize personalized drug selection. Recent topics of research using PDX have included the development of humanized mice as well as the use of 3D culture to complement some of the limitations of PDX models. CONCLUSIONS: UC PDX models serve as tools for understanding cancer biology, drug development and empowering precision medicine. The improvement of experimental systems using humanized mice to recapitulate the immune microenvironment of tumors will optimize UC PDX to study future questions in the field of immunotherapy.
Collapse
Affiliation(s)
- Yuki Kita
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Ryoichi Saito
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takahiro Inoue
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - William Y. Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Osamu Ogawa
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Kobayashi
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
106
|
Robertson N, Schook LB, Schachtschneider KM. Porcine cancer models: potential tools to enhance cancer drug trials. Expert Opin Drug Discov 2020; 15:893-902. [PMID: 32378979 DOI: 10.1080/17460441.2020.1757644] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The amount of time and money invested into cancer drug research, development, and clinical trials has continually increased over the past few decades. Despite record high cancer drug approval rates, cancer remains a leading cause of death. This suggests the need for more effective tools to help bring novel therapies to clinical practice in a timely manner. AREAS COVERED In this review, current issues associated with clinical trials are discussed, specifically focusing on poor accrual rates and time for trial completion. In addition, details regarding preclinical studies required before advancing to clinical trials are discussed, including advantages and limitations of current preclinical animal cancer models and their relevance to human cancer trials. Finally, new translational porcine cancer models (Oncopig Cancer Model (OCM)) are presented as potential co-clinical trial models. EXPERT OPINION In order to address issues impacting the poor success rate of oncology clinical trials, we propose the incorporation of the transformative OCM 'co-clinical trial' pathway into the cancer drug approval process. Due to the Oncopig's high homology to humans and similar tumor phenotypes, their utilization can provide improved preclinical prediction of both drug safety and efficacy prior to investing significant time and money in human clinical trials.
Collapse
Affiliation(s)
- Noah Robertson
- Department of Radiology, University of Illinois at Chicago , Chicago, IL, USA
| | - Lawrence B Schook
- Department of Radiology, University of Illinois at Chicago , Chicago, IL, USA.,Department of Animal Sciences, University of Illinois at Urbana-Champaign , Urbana, IL, USA
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago , Chicago, IL, USA.,Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago , Chicago, IL, USA
| |
Collapse
|
107
|
Liu LY, Ma XZ, Ouyang B, Ings DP, Marwah S, Liu J, Chen AY, Gupta R, Manuel J, Chen XC, Gage BK, Cirlan I, Khuu N, Chung S, Camat D, Cheng M, Sekhon M, Zagorovsky K, Abdou Mohamed MA, Thoeni C, Atif J, Echeverri J, Kollmann D, Fischer S, Bader GD, Chan WCW, Michalak TI, McGilvray ID, MacParland SA. Nanoparticle Uptake in a Spontaneous and Immunocompetent Woodchuck Liver Cancer Model. ACS NANO 2020; 14:4698-4715. [PMID: 32255624 DOI: 10.1021/acsnano.0c00468] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
There is a tremendous focus on the application of nanomaterials for the treatment of cancer. Nonprimate models are conventionally used to assess the biomedical utility of nanomaterials. However, these animals often lack an intact immunological background, and the tumors in these animals do not develop spontaneously. We introduce a preclinical woodchuck hepatitis virus-induced liver cancer model as a platform for nanoparticle (NP)-based in vivo experiments. Liver cancer development in these out-bred animals occurs as a result of persistent viral infection, mimicking human hepatitis B virus-induced HCC development. We highlight how this model addresses key gaps associated with other commonly used tumor models. We employed this model to (1) track organ biodistribution of gold NPs after intravenous administration, (2) examine their subcellular localization in the liver, (3) determine clearance kinetics, and (4) characterize the identity of hepatic macrophages that take up NPs using RNA-sequencing (RNA-seq). We found that the liver and spleen were the primary sites of NP accumulation. Subcellular analyses revealed accumulation of NPs in the lysosomes of CD14+ cells. Through RNA-seq, we uncovered that immunosuppressive macrophages within the woodchuck liver are the major cell type that take up injected NPs. The woodchuck-HCC model has the potential to be an invaluable tool to examine NP-based immune modifiers that promote host anti-tumor immunity.
Collapse
Affiliation(s)
- Lewis Y Liu
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- Department of Immunology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 6271, Toronto, Ontario, Canada M5S 1A8
| | - Xue-Zhong Ma
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Ben Ouyang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Rosebrugh Building, Room 407, Toronto, Ontario, Canada M5S 3G9
| | - Danielle P Ings
- Molecular Virology and Hepatology Research Group, Faculty of Medicine, Health Sciences Centre, Memorial University, 300 Prince Philip Drive, St. John's, Newfoundland, Canada A1B 3V6
| | - Sagar Marwah
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Jeff Liu
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Room 230, Toronto, Ontario, Canada M5S 3E1
| | - Annie Y Chen
- Molecular Virology and Hepatology Research Group, Faculty of Medicine, Health Sciences Centre, Memorial University, 300 Prince Philip Drive, St. John's, Newfoundland, Canada A1B 3V6
| | - Rahul Gupta
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Justin Manuel
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Xu-Chun Chen
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Blair K Gage
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- McEwen Stem Cell Institute, University Health Network, Toronto, Ontario, Canada M5G 1L7
| | - Iulia Cirlan
- Princess Margaret Genomics Centre, University Health Network, Toronto, Ontario, Canada M5G 1L7
| | - Nicholas Khuu
- Princess Margaret Genomics Centre, University Health Network, Toronto, Ontario, Canada M5G 1L7
| | - Sai Chung
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- Department of Immunology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 6271, Toronto, Ontario, Canada M5S 1A8
| | - Damra Camat
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- Department of Immunology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 6271, Toronto, Ontario, Canada M5S 1A8
| | - Michael Cheng
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- Luna Nanotech, Toronto, Ontario, Canada M5G 1Y8
| | - Manmeet Sekhon
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Kyryl Zagorovsky
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Rosebrugh Building, Room 407, Toronto, Ontario, Canada M5S 3G9
- Luna Nanotech, Toronto, Ontario, Canada M5G 1Y8
| | - Mohamed A Abdou Mohamed
- Luna Nanotech, Toronto, Ontario, Canada M5G 1Y8
- Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, Egypt 44519
| | - Cornelia Thoeni
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Jawairia Atif
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- Department of Immunology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 6271, Toronto, Ontario, Canada M5S 1A8
| | - Juan Echeverri
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Dagmar Kollmann
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Sandra Fischer
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 6271, Toronto, Ontario, Canada M5S 1A8
| | - Gary D Bader
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Room 230, Toronto, Ontario, Canada M5S 3E1
| | - Warren C W Chan
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Rosebrugh Building, Room 407, Toronto, Ontario, Canada M5S 3G9
- Department of Materials Science and Engineering, University of Toronto, 160 College Street, Room 450, Toronto, Ontario, Canada M5S 3E1
| | - Tomasz I Michalak
- Molecular Virology and Hepatology Research Group, Faculty of Medicine, Health Sciences Centre, Memorial University, 300 Prince Philip Drive, St. John's, Newfoundland, Canada A1B 3V6
| | - Ian D McGilvray
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Sonya A MacParland
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- Department of Immunology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 6271, Toronto, Ontario, Canada M5S 1A8
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 6271, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
108
|
Han K, Pierce SE, Li A, Spees K, Anderson GR, Seoane JA, Lo YH, Dubreuil M, Olivas M, Kamber RA, Wainberg M, Kostyrko K, Kelly MR, Yousefi M, Simpkins SW, Yao D, Lee K, Kuo CJ, Jackson PK, Sweet-Cordero A, Kundaje A, Gentles AJ, Curtis C, Winslow MM, Bassik MC. CRISPR screens in cancer spheroids identify 3D growth-specific vulnerabilities. Nature 2020; 580:136-141. [PMID: 32238925 PMCID: PMC7368463 DOI: 10.1038/s41586-020-2099-x] [Citation(s) in RCA: 180] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 01/10/2020] [Indexed: 12/18/2022]
Abstract
Cancer genomics studies have identified thousands of putative cancer driver genes1. Development of high-throughput and accurate models to define the functions of these genes is a major challenge. Here we devised a scalable cancer-spheroid model and performed genome-wide CRISPR screens in 2D monolayers and 3D lung-cancer spheroids. CRISPR phenotypes in 3D more accurately recapitulated those of in vivo tumours, and genes with differential sensitivities between 2D and 3D conditions were highly enriched for genes that are mutated in lung cancers. These analyses also revealed drivers that are essential for cancer growth in 3D and in vivo, but not in 2D. Notably, we found that carboxypeptidase D is responsible for removal of a C-terminal RKRR motif2 from the α-chain of the insulin-like growth factor 1 receptor that is critical for receptor activity. Carboxypeptidase D expression correlates with patient outcomes in patients with lung cancer, and loss of carboxypeptidase D reduced tumour growth. Our results reveal key differences between 2D and 3D cancer models, and establish a generalizable strategy for performing CRISPR screens in spheroids to reveal cancer vulnerabilities.
Collapse
Affiliation(s)
- Kyuho Han
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| | - Sarah E Pierce
- Program in Cancer Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Amy Li
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kaitlyn Spees
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Gray R Anderson
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jose A Seoane
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuan-Hung Lo
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Dubreuil
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Program in Cancer Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Micah Olivas
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Roarke A Kamber
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Wainberg
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Kaja Kostyrko
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Marcus R Kelly
- Program in Cancer Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Maryam Yousefi
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Scott W Simpkins
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - David Yao
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Keonil Lee
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Calvin J Kuo
- Program in Cancer Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Peter K Jackson
- Program in Cancer Biology, Stanford University School of Medicine, Stanford, CA, USA
- Baxter Laboratory, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Anshul Kundaje
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Andrew J Gentles
- Departments of Medicine and Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina Curtis
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Program in Cancer Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Program in Cancer Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael C Bassik
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Program in Cancer Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Program in Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA.
| |
Collapse
|
109
|
Development and characterization of mammary intraductal (MIND) spontaneous metastasis models for triple-negative breast cancer in syngeneic mice. Sci Rep 2020; 10:4681. [PMID: 32170125 PMCID: PMC7070052 DOI: 10.1038/s41598-020-61679-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/02/2020] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) has a more aggressive phenotype and higher metastasis and recurrence rates than other breast cancer subtypes. TNBC currently lacks a transplantation model that is suitable for clinical simulations of the tumor microenvironment. Intraductal injection of tumor cells into the mammary duct could mimic the occurrence and development of breast cancer. Herein, we injected 4T1 cells into the mammary ducts of BALB/C mice to build a preclinical model of TNBC and optimized the related construction method to observe the occurrence and spontaneous metastasis of tumors. We compared the effects of different cell numbers on tumorigenesis rates, times to tumorigenesis, and metastases to determine the optimal number of cells for modelling. We demonstrated that 4T1-MIND model mice injected with 20,000 cells revealed a suitable tumor formation rate and time, thus indicating a potential treatment time window after distant metastasis. We also injected 20,000 cells directly into the breast fat pad or breast duct for parallel comparison. The results still showed that the 4T1-MIND model provides sufficient treatment time for lung metastases in mice and that it is a more reliable model for early tumor development. The 4T1-MIND model requires continuous improvement and optimization. A suitable and optimized model for translational research and studies on the microenvironment in TNBC should be developed.
Collapse
|
110
|
Karyagina TS, Ulasov AV, Slastnikova TA, Rosenkranz AA, Lupanova TN, Khramtsov YV, Georgiev GP, Sobolev AS. Targeted Delivery of 111In Into the Nuclei of EGFR Overexpressing Cells via Modular Nanotransporters With Anti-EGFR Affibody. Front Pharmacol 2020; 11:176. [PMID: 32194412 PMCID: PMC7064642 DOI: 10.3389/fphar.2020.00176] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 02/07/2020] [Indexed: 12/18/2022] Open
Abstract
Since cell nucleus is one of the most vulnerable compartments, the maximum therapeutic effect from a variety of locally acting agents, such as photosensitizers, alfa-emitters, Auger electron emitters, will be expected when they get there. Therefore, the targeted delivery of these agents into the nuclei of target tumor cells is necessary for their anticancer effects and minimization of side effects. Modular nanotransporters (MNT) are artificial polypeptides comprising several predefined modules that recognize target cell, launching their subsequent internalization, escape from endosomes, and transport the drug load to the nucleus. This technology significantly enhances the cytotoxicity of locally acting drugs in vitro and in vivo. Epidermal growth factor receptors (EGFR) are useful molecular targets as they are overexpressed in glioblastoma, head-and-neck cancer, bladder cancer, and other malignancies. Here, we examined the possibility of using internalizable anti-EGFR affibody as an EGFR-targeting MNT module for drug transport into the cancer cell nuclei. It binds to both murine and human EGFR facilitating preclinical studies. We showed that MNT with affibody on the N-terminus (MNTN-affibody) effectively delivered the Auger electron emitter 111In to target cell nuclei and had pronounced cytotoxic efficacy against EGFR-overexpressing human A431 epidermoid carcinoma cells. Using EGFR-expressing human adenocarcinoma MCF-7 cells, we demonstrated that in contrast to MNT with N-terminal epidermal growth factor (EGF), MNTN-affibody and MNT with EGF on the C-terminus did not stimulate cancer cell proliferation.
Collapse
Affiliation(s)
- Tatiana S Karyagina
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexey V Ulasov
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Tatiana A Slastnikova
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrey A Rosenkranz
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.,Department of Biophysics, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Tatiana N Lupanova
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Yuri V Khramtsov
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Georgii P Georgiev
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexander S Sobolev
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.,Department of Biophysics, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
111
|
Partridge B, Rossmeisl JH. Companion animal models of neurological disease. J Neurosci Methods 2020; 331:108484. [PMID: 31733285 PMCID: PMC6942211 DOI: 10.1016/j.jneumeth.2019.108484] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 02/07/2023]
Abstract
Clinical translation of novel therapeutics that improve the survival and quality of life of patients with neurological disease remains a challenge, with many investigational drug and device candidates failing in advanced stage clinical trials. Naturally occurring inherited and acquired neurological diseases, such as epilepsy, inborn errors of metabolism, brain tumors, spinal cord injury, and stroke occur frequently in companion animals, and many of these share epidemiologic, pathophysiologic and clinical features with their human counterparts. As companion animals have a relatively abbreviated lifespan and genetic background, are immunocompetent, share their environment with human caregivers, and can be clinically managed using techniques and tools similar to those used in humans, they have tremendous potential for increasing the predictive value of preclinical drug and device studies. Here, we review comparative features of spontaneous neurological diseases in companion animals with an emphasis on neuroimaging methods and features, illustrate their historical use in translational studies, and discuss inherent limitations associated with each disease model. Integration of companion animals with naturally occurring disease into preclinical studies can complement and expand the knowledge gained from studies in other animal models, accelerate or improve the manner in which research is translated to the human clinic, and ultimately generate discoveries that will benefit the health of humans and animals.
Collapse
Affiliation(s)
- Brittanie Partridge
- Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA; Brain Tumor Center of Excellence, Wake Forest University Comprehensive Cancer Center, Medical Center Blvd, NRC 405, Winston Salem, NC, 27157, USA
| | - John H Rossmeisl
- Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA; Brain Tumor Center of Excellence, Wake Forest University Comprehensive Cancer Center, Medical Center Blvd, NRC 405, Winston Salem, NC, 27157, USA.
| |
Collapse
|
112
|
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
|
113
|
La transplantation de cellules tumorales chez le poisson zèbre : de la recherche translationnelle à la médecine personnalisée. Bull Cancer 2020; 107:30-40. [DOI: 10.1016/j.bulcan.2019.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/24/2022]
|
114
|
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
|
115
|
He H, Xu C, Cheng Z, Qian X, Zheng L. Drug Combinatorial Therapies for the Treatment of KRAS Mutated Lung Cancers. Curr Top Med Chem 2019; 19:2128-2142. [PMID: 31475900 DOI: 10.2174/1568026619666190902150555] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/23/2019] [Accepted: 07/04/2019] [Indexed: 02/08/2023]
Abstract
KRAS is the most common oncogene to be mutated in lung cancer, and therapeutics directly targeting KRAS have proven to be challenging. The mutations of KRAS are associated with poor prognosis, and resistance to both adjuvant therapy and targeted EGFR TKI. EGFR TKIs provide significant clinical benefit for patients whose tumors bear EGFR mutations. However, tumors with KRAS mutations rarely respond to the EGFR TKI therapy. Thus, combination therapy is essential for the treatment of lung cancers with KRAS mutations. EGFR TKI combined with inhibitors of MAPKs, PI3K/mTOR, HDAC, Wee1, PARP, CDK and Hsp90, even miRNAs and immunotherapy, were reviewed. Although the effects of the combination vary, the combined therapeutics are one of the best options at present to treat KRAS mutant lung cancer.
Collapse
Affiliation(s)
- Hao He
- School of Pharmacy, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Chang Xu
- National Vaccine & Serum Institute, Beijing, China
| | - Zhao Cheng
- School of Pharmacy, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Xiaoying Qian
- School of Pharmacy, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Lei Zheng
- School of Pharmacy, Xi'an Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
116
|
Castle KD, Kirsch DG. Establishing the Impact of Vascular Damage on Tumor Response to High-Dose Radiation Therapy. Cancer Res 2019; 79:5685-5692. [PMID: 31427377 PMCID: PMC6948140 DOI: 10.1158/0008-5472.can-19-1323] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/17/2019] [Accepted: 08/07/2019] [Indexed: 12/26/2022]
Abstract
Approximately half of all patients with cancer receive radiotherapy, which is conventionally delivered in relatively small doses (1.8-2 Gy) per daily fraction over one to two months. Stereotactic body radiation therapy (SBRT), in which a high daily radiation dose is delivered in 1 to 5 fractions, has improved local control rates for several cancers. However, despite the widespread adoption of SBRT in the clinic, controversy surrounds the mechanism by which SBRT enhances local control. Some studies suggest that high doses of radiation (≥10 Gy) trigger tumor endothelial cell death, resulting in indirect killing of tumor cells through nutrient depletion. On the other hand, mathematical models predict that the high radiation dose per fraction used in SBRT increases direct tumor cell killing, suggesting that disruption of the tumor vasculature is not a critical mediator of tumor cure. Here, we review the application of genetically engineered mouse models to radiosensitize tumor cells or endothelial cells to dissect the role of these cellular targets in mediating the response of primary tumors to high-dose radiotherapy in vivo These studies demonstrate a role for endothelial cell death in mediating tumor growth delay, but not local control following SBRT.
Collapse
Affiliation(s)
- Katherine D Castle
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - David G Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina.
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
117
|
Overgaard NH, Fan TM, Schachtschneider KM, Principe DR, Schook LB, Jungersen G. Of Mice, Dogs, Pigs, and Men: Choosing the Appropriate Model for Immuno-Oncology Research. ILAR J 2019; 59:247-262. [PMID: 30476148 DOI: 10.1093/ilar/ily014] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 07/30/2018] [Indexed: 02/06/2023] Open
Abstract
The immune system plays dual roles in response to cancer. The host immune system protects against tumor formation via immunosurveillance; however, recognition of the tumor by immune cells also induces sculpting mechanisms leading to a Darwinian selection of tumor cell variants with reduced immunogenicity. Cancer immunoediting is the concept used to describe the complex interplay between tumor cells and the immune system. This concept, commonly referred to as the three E's, is encompassed by 3 distinct phases of elimination, equilibrium, and escape. Despite impressive results in the clinic, cancer immunotherapy still has room for improvement as many patients remain unresponsive to therapy. Moreover, many of the preclinical results obtained in the widely used mouse models of cancer are lost in translation to human patients. To improve the success rate of immuno-oncology research and preclinical testing of immune-based anticancer therapies, using alternative animal models more closely related to humans is a promising approach. Here, we describe 2 of the major alternative model systems: canine (spontaneous) and porcine (experimental) cancer models. Although dogs display a high rate of spontaneous tumor formation, an increased number of genetically modified porcine models exist. We suggest that the optimal immuno-oncology model may depend on the stage of cancer immunoediting in question. In particular, the spontaneous canine tumor models provide a unique platform for evaluating therapies aimed at the escape phase of cancer, while genetically engineered swine allow for elucidation of tumor-immune cell interactions especially during the phases of elimination and equilibrium.
Collapse
Affiliation(s)
- Nana H Overgaard
- Department of Micro- and Nanotechnology, Technical University of Denmark, Kgs Lyngby, Denmark
| | - Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana-Champaign, Illinois
| | | | - Daniel R Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, Illinois
| | - Lawrence B Schook
- Department of Radiology, University of Illinois, Chicago, Illinois.,Department of Animal Sciences, University of Illinois, Urbana-Champaign, Illinois
| | - Gregers Jungersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
118
|
Sieber S, Grossen P, Bussmann J, Campbell F, Kros A, Witzigmann D, Huwyler J. Zebrafish as a preclinical in vivo screening model for nanomedicines. Adv Drug Deliv Rev 2019; 151-152:152-168. [PMID: 30615917 DOI: 10.1016/j.addr.2019.01.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/23/2018] [Accepted: 01/02/2019] [Indexed: 12/11/2022]
Abstract
The interactions of nanomedicines with biological environments is heavily influenced by their physicochemical properties. Formulation design and optimization are therefore key steps towards successful nanomedicine development. Unfortunately, detailed assessment of nanomedicine formulations, at a macromolecular level, in rodents is severely limited by the restricted imaging possibilities within these animals. Moreover, rodent in vivo studies are time consuming and expensive, limiting the number of formulations that can be practically assessed in any one study. Consequently, screening and optimisation of nanomedicine formulations is most commonly performed in surrogate biological model systems, such as human-derived cell cultures. However, despite the time and cost advantages of classical in vitro models, these artificial systems fail to reflect and mimic the complex biological situation a nanomedicine will encounter in vivo. This has acutely hampered the selection of potentially successful nanomedicines for subsequent rodent in vivo studies. Recently, zebrafish have emerged as a promising in vivo model, within nanomedicine development pipelines, by offering opportunities to quickly screen nanomedicines under in vivo conditions and in a cost-effective manner so as to bridge the current gap between in vitro and rodent studies. In this review, we outline several advantageous features of the zebrafish model, such as biological conservation, imaging modalities, availability of genetic tools and disease models, as well as their various applications in nanomedicine development. Critical experimental parameters are discussed and the most beneficial applications of the zebrafish model, in the context of nanomedicine development, are highlighted.
Collapse
Affiliation(s)
- Sandro Sieber
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Philip Grossen
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Jeroen Bussmann
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Frederick Campbell
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Alexander Kros
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland; Department of Biochemistry and Molecular Biology, University of British Columbia, Health Sciences Mall, Vancouver, British Columbia, Canada..
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| |
Collapse
|
119
|
Houghton PJ, Kurmasheva RT. Challenges and Opportunities for Childhood Cancer Drug Development. Pharmacol Rev 2019; 71:671-697. [PMID: 31558580 PMCID: PMC6768308 DOI: 10.1124/pr.118.016972] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer in children is rare with approximately 15,700 new cases diagnosed in the United States annually. Through use of multimodality therapy (surgery, radiation therapy, and aggressive chemotherapy), 70% of patients will be "cured" of their disease, and 5-year event-free survival exceeds 80%. However, for patients surviving their malignancy, therapy-related long-term adverse effects are severe, with an estimated 50% having chronic life-threatening toxicities related to therapy in their fourth or fifth decade of life. While overall intensive therapy with cytotoxic agents continues to reduce cancer-related mortality, new understanding of the molecular etiology of many childhood cancers offers an opportunity to redirect efforts to develop effective, less genotoxic therapeutic options, including agents that target oncogenic drivers directly, and the potential for use of agents that target the tumor microenvironment and immune-directed therapies. However, for many high-risk cancers, significant challenges remain.
Collapse
Affiliation(s)
- Peter J Houghton
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| | - Raushan T Kurmasheva
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| |
Collapse
|
120
|
Shi C, Chen X, Tan D. Development of patient-derived xenograft models of prostate cancer for maintaining tumor heterogeneity. Transl Androl Urol 2019; 8:519-528. [PMID: 31807428 DOI: 10.21037/tau.2019.08.31] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Prostate cancer (Pca) is a heterogeneous disease with multiple morphological patterns. Thus, the establishment of a patient-derived xenograft (PDX) model that retains key features of the primary tumor is of great significance. This review demonstrates the characteristics and advantages of the Pca PDX model and summarizes the main factors affecting the establishment of the model. Because this model well recapitulates the diverse heterogeneity observed in the clinic, it was extensively utilized to discover new therapeutic targets, screen drugs, and explore metastatic mechanisms. In the future, clinical phenotype and different stages of the Pca patient might be faithfully reflected by PDX model, which provides tremendous potential for understanding Pca biology and achieving individualized treatment.
Collapse
Affiliation(s)
- Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China.,School of Basic Medical Sciences, the Chengdu Medical University, Xindu 610500, China
| | - Xue Chen
- Division of Cancer Biology, Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China.,School of Basic Medical Sciences, the Chengdu Medical University, Xindu 610500, China
| | - Dengxu Tan
- Division of Cancer Biology, Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
121
|
Nigjeh SE, Yeap SK, Nordin N, Rahman H, Rosli R. In Vivo Anti-Tumor Effects of Citral on 4T1 Breast Cancer Cells via Induction of Apoptosis and Downregulation of Aldehyde Dehydrogenase Activity. Molecules 2019; 24:molecules24183241. [PMID: 31492037 PMCID: PMC6767168 DOI: 10.3390/molecules24183241] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer and the leading cause of cancer death among females globally. The tumorigenic activities of cancer cells such as aldehyde dehydrogenase (ALDH) activity and differentiation have contributed to relapse and eventual mortality in breast cancer. Thus, current drug discovery research is focused on targeting breast cancer cells with ALDH activity and their capacity to form secondary tumors. Citral (3,7-dimethyl-2,6-octadienal), from lemon grass (Cymbopogoncitrates), has been previously reported to have a cytotoxic effect on breast cancer cells. Hence, this study was conducted to evaluate the in vivo effect of citral in targeting ALDH activity of breast cancer cells. BALB/c mice were challenged with 4T1 breast cancer cells followed by daily oral feeding of 50 mg/kg citral or distilled water for two weeks. The population of ALDH+ tumor cells and their capacity to form secondary tumors in both untreated and citral treated 4T1 challenged mice were assessed by Aldefluor assay and tumor growth upon cell reimplantation in normal mice, respectively. Citral treatment reduced the size and number of cells with ALDH+ activity of the tumors in 4T1-challenged BALB/c mice. Moreover, citral-treated mice were also observed with smaller tumor size and delayed tumorigenicity after reimplantation of the primary tumor cells into normal mice. These findings support the antitumor effect of citral in targeting ALDH+ cells and tumor recurrence in breast cancer cells.
Collapse
Affiliation(s)
- Siyamak Ebrahimi Nigjeh
- Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Faculty of Life Science and Biotechnology, Shahid Beheshti University, Daneshjou Boulevard, Tehran 1983969411, Iran
- Department of Medical Genetics, Tehran University of Medical Sciences, Poursina street, Tehran 1366736511, Iran
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Jalan Sunsuria, Bandar Sunsuria, Sepang 43900, Selangor, Malaysia
| | - Norshariza Nordin
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Heshu Rahman
- Department of Medical Laboratory Sciences and Technology, College of Health Sciences, Komar University of Science and Technology, Chaq Chaq Qularaese, Sarchinar District, Sulaimani 334, Iraq
- Department of Clinical and Internal Medicine, College of Veterinary Medicine, University of Sulaimani, Sulaimani 334, Iraq
| | - Rozita Rosli
- Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| |
Collapse
|
122
|
Faithful preclinical mouse models for better translation to bedside in the field of immuno-oncology. Int J Clin Oncol 2019; 25:831-841. [PMID: 31407168 DOI: 10.1007/s10147-019-01520-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/01/2019] [Indexed: 12/19/2022]
Abstract
The success of immunotherapy using immune checkpoint inhibitors has changed the practice of cancer treatment tremendously. However, there are still many clinical challenges, such as drug resistance, predictive biomarker development, exploration of combination therapies, and prediction of immune-related adverse events in preclinical settings. To overcome these problems, it is essential to establish faithful preclinical mouse models that recapitulate the clinical features, molecular genetics, biological heterogeneity, and immune microenvironment of human cancers. Here we review the advantages and disadvantages of current preclinical mouse models, including syngeneic murine tumor cell lines, autochthonous tumor models, cancer cell line-derived xenografts, patient-derived-xenografts, and various kinds of immunologically humanized mice. We discuss how these models should be characterized and applied in preclinical settings, and how we should prepare preclinical studies for successful translation from bench to bedside.
Collapse
|
123
|
Willers C, Svitina H, Rossouw MJ, Swanepoel RA, Hamman JH, Gouws C. Models used to screen for the treatment of multidrug resistant cancer facilitated by transporter-based efflux. J Cancer Res Clin Oncol 2019; 145:1949-1976. [PMID: 31292714 DOI: 10.1007/s00432-019-02973-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/04/2019] [Indexed: 01/09/2023]
Abstract
PURPOSE Efflux transporters of the adenosine triphosphate-binding cassette (ABC)-superfamily play an important role in the development of multidrug resistance (multidrug resistant; MDR) in cancer. The overexpression of these transporters can directly contribute to the failure of chemotherapeutic drugs. Several in vitro and in vivo models exist to screen for the efficacy of chemotherapeutic drugs against MDR cancer, specifically facilitated by efflux transporters. RESULTS This article reviews a range of efflux transporter-based MDR models used to test the efficacy of compounds to overcome MDR in cancer. These models are classified as either in vitro or in vivo and are further categorised as the most basic, conventional models or more complex and advanced systems. Each model's origin, advantages and limitations, as well as specific efflux transporter-based MDR applications are discussed. Accordingly, future modifications to existing models or new research approaches are suggested to develop prototypes that closely resemble the true nature of multidrug resistant cancer in the human body. CONCLUSIONS It is evident from this review that a combination of both in vitro and in vivo preclinical models can provide a better understanding of cancer itself, than using a single model only. However, there is still a clear lack of progression of these models from basic research to high-throughput clinical practice.
Collapse
Affiliation(s)
- Clarissa Willers
- Pharmacen™, Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - Hanna Svitina
- Pharmacen™, Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - Michael J Rossouw
- Pharmacen™, Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - Roan A Swanepoel
- Pharmacen™, Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - Josias H Hamman
- Pharmacen™, Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - Chrisna Gouws
- Pharmacen™, Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa.
| |
Collapse
|
124
|
McVeigh LE, Wijetunga I, Ingram N, Marston G, Prasad R, Markham AF, Coletta PL. Development of orthotopic tumour models using ultrasound-guided intrahepatic injection. Sci Rep 2019; 9:9904. [PMID: 31289364 PMCID: PMC6616610 DOI: 10.1038/s41598-019-46410-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/25/2019] [Indexed: 01/19/2023] Open
Abstract
Mouse models of human diseases are an essential part of the translational pipeline. Orthotopic tumour mouse models are increasingly being used in cancer research due to their increased clinical relevance over subcutaneous xenograft models, particularly in relation to metastatic disease. In this study, we have developed orthotopic colorectal cancer liver metastases (CRCLM) and primary cholangiocarcinoma (CCA) models in BALB/c nude mice using minimally invasive ultrasound-guided intrahepatic injection. Due to its minimally invasive nature, the method reduced risk from surgical complications whilst being fast and easy to perform and resulted in measurable tumour volumes 1 to 3 weeks post-injection. Tumour volumes were monitored in vivo by weekly high-frequency ultrasound (HF-US) and/or twice weekly bioluminescence imaging (BLI) and confirmed with end-point histology. Take rates were high for human CRC cells (>73%) and for CCA cells (90%). We have demonstrated that this method reliably induces CRCLM and CCAs, in which tumour volume can be monitored throughout using HF-US and/or BLI. This provides a promising experimental tool for future testing of cancer therapeutics in an orthotopic model.
Collapse
Affiliation(s)
- L E McVeigh
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, LS9 7TF, UK.
| | - I Wijetunga
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, LS9 7TF, UK
| | - N Ingram
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, LS9 7TF, UK
| | - G Marston
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, LS9 7TF, UK
| | - R Prasad
- Department of Hepatobiliary and Transplant Surgery, St. James's University Hospital, Leeds, LS9 7TF, UK
| | - A F Markham
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, LS9 7TF, UK
| | - P L Coletta
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, LS9 7TF, UK
| |
Collapse
|
125
|
Pfefferle AD, Darr DB, Calhoun BC, Mott KR, Rosen JM, Perou CM. The MMTV-Wnt1 murine model produces two phenotypically distinct subtypes of mammary tumors with unique therapeutic responses to an EGFR inhibitor. Dis Model Mech 2019; 12:dmm.037192. [PMID: 31213486 PMCID: PMC6679375 DOI: 10.1242/dmm.037192] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 06/06/2019] [Indexed: 12/23/2022] Open
Abstract
The Wnt gene family encodes an evolutionarily conserved group of proteins that regulate cell growth, differentiation and stem cell self-renewal. Aberrant Wnt signaling in human breast tumors has been proposed as a driver of tumorigenesis, especially in the basal-like tumor subtype where canonical Wnt signaling is both enriched and predictive of poor clinical outcomes. The development of effective Wnt-based therapeutics, however, has been slowed in part by a limited understanding of the context-dependent nature with which these aberrations influence breast tumorigenesis. We previously reported that MMTV-Wnt1 mice, an established model for studying Wnt signaling in breast tumors, develop two subtypes of tumors by gene expression classification: Wnt1-EarlyEx and Wnt1-LateEx Here, we extend this initial observation and show that Wnt1-EarlyEx tumors exhibit high expression of canonical Wnt, non-canonical Wnt, and EGFR signaling pathway signatures. Therapeutically, Wnt1-EarlyEx tumors showed a dynamic reduction in tumor volume when treated with an EGFR inhibitor. Wnt1-EarlyEx tumors had primarily Cd49fpos/Epcamneg FACS profiles, but it was not possible to serially transplant these tumors into wild-type FVB female mice. Conversely, Wnt1-LateEx tumors had a bloody gross pathology, which was highlighted by the presence of 'blood lakes' identified by H&E staining. These tumors had primarily Cd49fpos/Epcampos FACS profiles, but also contained a secondary Cd49fpos/Epcamneg subpopulation. Wnt1-LateEx tumors were enriched for activating Hras1 mutations and were capable of reproducing tumors when serially transplanted into wild-type FVB female mice. This study definitively shows that the MMTV-Wnt1 mouse model produces two phenotypically distinct subtypes of mammary tumors that differ in multiple biological aspects including sensitivity to an EGFR inhibitor.
Collapse
Affiliation(s)
- Adam D Pfefferle
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - David B Darr
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Benjamin C Calhoun
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kevin R Mott
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.,Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Charles M Perou
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA .,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.,Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
126
|
Malcolm JE, Stearns TM, Airhart SD, Graber JH, Bult CJ. Factors that influence response classifications in chemotherapy treated patient-derived xenografts (PDX). PeerJ 2019; 7:e6586. [PMID: 30944774 PMCID: PMC6441558 DOI: 10.7717/peerj.6586] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 02/08/2019] [Indexed: 01/06/2023] Open
Abstract
In this study, we investigated the impact of initial tumor volume, rate of tumor growth, cohort size, study duration, and data analysis method on chemotherapy treatment response classifications in patient-derived xenografts (PDXs). The analyses were conducted on cisplatin treatment response data for 70 PDX models representing ten cancer types with up to 28-day study duration and cohort sizes of 3-10 tumor-bearing mice. The results demonstrated that a 21-day dosing study using a cohort size of eight was necessary to reliably detect responsive models (i.e., tumor volume ratio of treated animals to control between 0.1 and 0.42)-independent of analysis method. A cohort of three tumor-bearing animals led to a reliable classification of models that were both highly responsive and highly nonresponsive to cisplatin (i.e., tumor volume ratio of treated animals to control animals less than 0.10). In our set of PDXs, we found that tumor growth rate in the control group impacted treatment response classification more than initial tumor volume. We repeated the study design factors using docetaxel treated PDXs with consistent results. Our results highlight the importance of defining endpoints for PDX dosing studies when deciding the size of cohorts to use in dosing studies and illustrate that response classifications for a study do not differ significantly across the commonly used analysis methods that are based on tumor volume changes in treatment versus control groups.
Collapse
Affiliation(s)
- Joan E. Malcolm
- The Jackson Laboratory, Bar Harbor, ME, United States of America
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States of America
| | | | - Susan D. Airhart
- The Jackson Laboratory, Bar Harbor, ME, United States of America
| | - Joel H. Graber
- The Jackson Laboratory, Bar Harbor, ME, United States of America
- The MDI Biological Laboratory, Bar Harbor, ME, United States of America
| | - Carol J. Bult
- The Jackson Laboratory, Bar Harbor, ME, United States of America
| |
Collapse
|
127
|
Ward C, Meehan J, Gray M, Kunkler IH, Langdon SP, Murray A, Argyle D. Preclinical Organotypic Models for the Assessment of Novel Cancer Therapeutics and Treatment. Curr Top Microbiol Immunol 2019. [PMID: 30859401 DOI: 10.1007/82_2019_159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The immense costs in both financial terms and preclinical research effort that occur in the development of anticancer drugs are unfortunately not matched by a substantial increase in improved clinical therapies due to the high rate of failure during clinical trials. This may be due to issues with toxicity or lack of clinical effectiveness when the drug is evaluated in patients. Currently, much cancer research is driven by the need to develop therapies that can exploit cancer cell adaptations to conditions in the tumor microenvironment such as acidosis and hypoxia, the requirement for more-specific, targeted treatments, or the exploitation of 'precision medicine' that can target known genomic changes in patient DNA. The high attrition rate for novel anticancer therapies suggests that the preclinical methods used in screening anticancer drugs need improvement. This chapter considers the advantages and disadvantages of 3D organotypic models in both cancer research and cancer drug screening, particularly in the areas of targeted drugs and the exploitation of genomic changes that can be used for therapeutic advantage in precision medicine.
Collapse
Affiliation(s)
- Carol Ward
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Roslin, Midlothian, EH25 9RG, Edinburgh, UK.
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU, Edinburgh, UK.
| | - James Meehan
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU, Edinburgh, UK
- School of Engineering and Physical Sciences, Institute of Sensors, Signals and Systems, Heriot-Watt University, EH14 4AS, Edinburgh, UK
| | - Mark Gray
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Roslin, Midlothian, EH25 9RG, Edinburgh, UK
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU, Edinburgh, UK
| | - Ian H Kunkler
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU, Edinburgh, UK
| | - Simon P Langdon
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU, Edinburgh, UK
| | - Alan Murray
- School of Engineering, Faraday Building, The King's Buildings, Mayfield Road, EH9 3JL, Edinburgh, UK
| | - David Argyle
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Roslin, Midlothian, EH25 9RG, Edinburgh, UK
| |
Collapse
|
128
|
Dhiman N, Kingshott P, Sumer H, Sharma CS, Rath SN. On-chip anticancer drug screening - Recent progress in microfluidic platforms to address challenges in chemotherapy. Biosens Bioelectron 2019; 137:236-254. [PMID: 31121461 DOI: 10.1016/j.bios.2019.02.070] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 12/18/2022]
Abstract
There is an increasing need for advanced and inexpensive preclinical models to accelerate the development of anticancer drugs. While costly animal models fail to predict human clinical outcomes, in vitro models such as microfluidic chips ('tumor-on-chip') are showing tremendous promise at predicting and providing meaningful preclinical drug screening outcomes. Research on 'tumor-on-chips' has grown enormously worldwide and is being widely accepted by pharmaceutical companies as a drug development tool. In light of this shift in philosophy, it is important to review the recent literature on microfluidic devices to determine how rapidly the technology has progressed as a promising model for drug screening and aiding cancer therapy. We review the past five years of successful developments and capabilities in microdevice technology (cancer models) for use in anticancer drug screening. Microfluidic devices that are being designed to address current challenges in chemotherapy, such as drug resistance, combinatorial drug therapy, personalized medicine, and cancer metastasis are also reviewed in detail. We provide a perspective on how personalized 'tumor-on-chip', as well as high-throughput microfluidic platforms based on patient-specific tumor cells, can potentially replace the more expensive and 'non-human' animal models in preclinical anticancer drug development.
Collapse
Affiliation(s)
- Nandini Dhiman
- Regenerative Medicine and Stem Cells Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India; Department of Chemistry and Biotechnology, Faculty of Science and Engineering Technology, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Peter Kingshott
- Department of Chemistry and Biotechnology, Faculty of Science and Engineering Technology, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Huseyin Sumer
- Department of Chemistry and Biotechnology, Faculty of Science and Engineering Technology, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Chandra S Sharma
- Creative & Advanced Research Based On Nanomaterials Laboratory, Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Subha Narayan Rath
- Regenerative Medicine and Stem Cells Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India.
| |
Collapse
|
129
|
Bailey KL, Carlson MA. Porcine Models of Pancreatic Cancer. Front Oncol 2019; 9:144. [PMID: 30915276 PMCID: PMC6423062 DOI: 10.3389/fonc.2019.00144] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/20/2019] [Indexed: 01/29/2023] Open
Abstract
Pancreatic cancer is the fourth most common cause of cancer-related deaths in both men and women. The 5-year survival rate for metastatic pancreatic cancer is only 8%. There remains a need for improved early diagnosis and therapy for pancreatic cancer. Murine models are the current standard for preclinical study of pancreatic cancer. However, mice may not accurately reflect human biology because of a variety of differences between the two species. Remarkably, only 5-8% of anti-cancer drugs that have emerged from preclinical studies and entered clinical studies have ultimately been approved for clinical use. The cause of this poor approval rate is multi-factorial, but may in part be due to use of murine models that have limited accuracy with respect to human disease. Murine models also have limited utility in the development of diagnostic or interventional technology that require a human-sized model. So, at present, there remains a need for improved animal models of pancreatic cancer. The rationale for a porcine model of pancreatic cancer is (i) to enable development of diagnostic/therapeutic devices for which murine models have limited utility; and (ii) to have a highly predictive preclinical model in which anti-cancer therapies can be tested and optimized prior to a clinical trial. Recently, pancreatic tumors were induced in transgenic Oncopigs and porcine pancreatic ductal cells were transformed that contain oncogenic KRAS and p53-null mutations. Both techniques to induce pancreatic tumors in pigs are undergoing further refinement and expansion. The Oncopig currently is commercially available, and it is conceivable that other porcine models of pancreatic cancer may be available for general use in the near future.
Collapse
Affiliation(s)
- Katie L. Bailey
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mark A. Carlson
- Department of Surgery and Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States,Department of Surgery, VA Nebraska-Western Iowa Health Care System, Omaha, NE, United States,*Correspondence: Mark A. Carlson
| |
Collapse
|
130
|
O'Loughlin TA, Gilbert LA. Functional Genomics for Cancer Research: Applications In Vivo and In Vitro. ANNUAL REVIEW OF CANCER BIOLOGY 2019. [DOI: 10.1146/annurev-cancerbio-030518-055742] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Functional genomics holds great promise for the dissection of cancer biology. The elucidation of genetic cooperation and molecular details that govern oncogenesis, metastasis, and response to therapy is made possible by robust technologies for perturbing gene function coupled to quantitative analysis of cancer phenotypes resulting from genetic or epigenetic perturbations. Multiplexed genetic perturbations enable the dissection of cooperative genetic lesions as well as the identification of synthetic lethal gene pairs that hold particular promise for constructing innovative cancer therapies. Lastly, functional genomics strategies enable the highly multiplexed in vivo analysis of genes that govern tumorigenesis as well as of the complex multicellular biology of a tumor, such as immune response and metastasis phenotypes. In this review, we discuss both historical and emerging functional genomics approaches and their impact on the cancer research landscape.
Collapse
Affiliation(s)
- Thomas A. O'Loughlin
- Department of Urology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94158, USA
| | - Luke A. Gilbert
- Department of Urology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94158, USA
- Innovative Genomics Institute, University of California, San Francisco, California 94158, USA
| |
Collapse
|
131
|
A step towards valid detection and quantification of lung cancer volume in experimental mice with contrast agent-based X-ray microtomography. Sci Rep 2019; 9:1325. [PMID: 30718557 PMCID: PMC6362109 DOI: 10.1038/s41598-018-37394-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 11/30/2018] [Indexed: 12/18/2022] Open
Abstract
Tumor volume is a parameter used to evaluate the performance of new therapies in lung cancer research. Conventional methods that are used to estimate tumor size in mouse models fail to provide fast and reliable volumetric data for tumors grown non-subcutaneously. Here, we evaluated the use of iodine-staining combined with micro-computed tomography (micro-CT) to estimate the tumor volume of ex vivo tumor-burdened lungs. We obtained fast high spatial resolution three-dimensional information of the lungs, and we demonstrated that iodine-staining highlights tumors and unhealthy tissue. We processed iodine-stained lungs for histopathological analysis with routine hematoxylin and eosin (H&E) staining. We compared the traditional tumor burden estimation performed manually with H&E histological slices with a semi-automated method using micro-CT datasets. In mouse models that develop lung tumors with well precise boundaries, the method that we describe here enables to perform a quick estimation of tumorous tissue volume in micro-CT images. Our method overestimates the tumor burden in tumors surrounded by abnormal tissue, while traditional histopathological analysis underestimates tumor volume. We propose to embed micro-CT imaging to the traditional workflow of tumorous lung analyses in preclinical cancer research as a strategy to obtain a more accurate estimation of the total lung tumor burden.
Collapse
|
132
|
Akimoto J, Nakayama M, Takagi S, Okano T. Efficient intrahepatic tumor generation by cell sheet transplantation to fabricate orthotopic hepatocarcinoma-bearing model mice for drug testing. J Biomed Mater Res A 2019; 107:1071-1079. [PMID: 30706672 DOI: 10.1002/jbm.a.36641] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 01/24/2019] [Indexed: 11/10/2022]
Abstract
Subcutaneous tumor-bearing mice are commonly used to evaluate antitumor activity in preclinical studies of anticancer drugs. However, these models often exhibit excessive antitumor responses to anticancer drug candidates. In this study, intrahepatic tumor-bearing mice as orthotopic tumor models were fabricated by transplanting hepatocarcinoma cell monolayers (sheets) to investigate differences in ectopic versus orthotopic antitumor response. Cell sheets, harvested from temperature-responsive cell culture dishes using thin gelatin gel supporters, were transferred onto mouse liver surfaces. Cell sheet transplantation drastically improved intrahepatic tumor formation compared with direct intrahepatic injection of dispersed cells. In particular, all cell sheet-transplanted mice formed well-developed tumors inside the liver following removal of the mesothelial membrane at the liver surface. Notably, these mice exhibited comparable life spans, indicating similar intrahepatic tumor development rates. Antitumor activity of doxorubicin (DOX) was examined using both subcutaneous and intrahepatic tumor-bearing mice. Although DOX administration yielded decreased subcutaneous tumor volumes, intrahepatic tumors exhibited no significant antitumor response. The results were considered to represent pharmacokinetic and histological structure differences between ectopic and orthotopic tumors, and partially supported the clinical uses of DOX. Therefore, cancer cell sheet transplantation constitutes a promising method to fabricate intrahepatic tumor-bearing mice for drug screening test in preclinical studies. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1071-1079, 2019.
Collapse
Affiliation(s)
- Jun Akimoto
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Kawada-cho 8-1, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Masamichi Nakayama
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Kawada-cho 8-1, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Soichi Takagi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Kawada-cho 8-1, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Kawada-cho 8-1, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
133
|
Jones M, Bellusci S. Imaging and Analysis of Mouse Embryonic Whole Lung, Isolated Tissue, and Lineage-Labelled Cell Culture. Methods Mol Biol 2019; 1940:109-127. [PMID: 30788821 DOI: 10.1007/978-1-4939-9086-3_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Research on lung development and disease frequently utilizes mouse models to conduct in vitro experiments. Such experiments involve multiple methodologically distinct stages, from careful consideration of mouse models used to obtain biological samples, to the culturing and imaging of those samples, and finally, to post-imaging analysis. Here, we detail basic protocols to assist with each of these stages. First, we discuss harvesting and preparing biological samples; second, we focus on culturing embryonic whole lung explants and isolated mesenchyme and epithelium; third, we specify the basics of obtaining still and live images; and finally, we bring these methods together by considering and briefly analyzing a lineage-labelling experiment.
Collapse
Affiliation(s)
- Matthew Jones
- Faculty of Medicine, Excellence Cluster Cardio-Pulmonary System (ECCPS), University of Giessen, Giessen, Germany
| | - Saverio Bellusci
- Faculty of Medicine, Excellence Cluster Cardio-Pulmonary System (ECCPS), University of Giessen, Giessen, Germany.
| |
Collapse
|
134
|
Zhou K, Ding Y, Vuletic I, Tian Y, Li J, Liu J, Huang Y, Sun H, Li C, Ren Q, Lu Y. In vivo long-term investigation of tumor bearing mKate2 by an in-house fluorescence molecular imaging system. Biomed Eng Online 2018; 17:187. [PMID: 30594200 PMCID: PMC6310933 DOI: 10.1186/s12938-018-0615-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/05/2018] [Indexed: 11/10/2022] Open
Abstract
Background Optical imaging is one of the most common, low-cost imaging tools used for investigating the tumor biological behavior in vivo. This study explores the feasibility and sensitivity of a near infrared fluorescent protein mKate2 for a long-term non-invasive tumor imaging in BALB/c nude mice, by using a low-power optical imaging system. Methods In this study, breast cancer cell line MDA-MB-435s expressing mKate2 and MDA-MB-231 expressing a dual reporter gene firefly luciferase (fLuc)-GFP were used as cell models. Tumor cells were implanted in different animal body compartments including subcutaneous, abdominal and deep tissue area and closely monitored in real-time. A simple and low-power optical imaging system was set up to image both fluorescence and bioluminescence in live animals. Results The presence of malignant tissue was further confirmed by histopathological assay. Considering its lower exposure time and no need of substrate injection, mKate2 is considered a superior choice for subcutaneous imaging compared with fLuc. On the contrary, fLuc has shown to be a better option when monitoring the tumor in a diffusive area such as abdominal cavity. Furthermore, both reporter genes have shown good stability and sensitivity for deep tissue imaging, i.e. tumor within the liver. In addition, fLuc has shown to be an excellent method for detecting tumor cells in the lung. Conclusions The combination of mKate2 and fLuc offers a superior choice for long-term non-invasive real-time investigation of tumor biological behavior in vivo.
Collapse
Affiliation(s)
- Kedi Zhou
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Beijing, 100871, China
| | - Yichen Ding
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Beijing, 100871, China
| | - Ivan Vuletic
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Beijing, 100871, China
| | - Yonglu Tian
- Laboratory Animal Centre, Peking University, No. 5 Yiheyuan Road, Beijing, 100871, China
| | - Jun Li
- Laboratory Animal Centre, Peking University, No. 5 Yiheyuan Road, Beijing, 100871, China
| | - Jinghao Liu
- Laboratory Animal Centre, Peking University, No. 5 Yiheyuan Road, Beijing, 100871, China
| | - Yixing Huang
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Beijing, 100871, China
| | - Hongfang Sun
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Beijing, 100871, China.
| | - Changhui Li
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Beijing, 100871, China
| | - Qiushi Ren
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Beijing, 100871, China
| | - Yanye Lu
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Beijing, 100871, China.
| |
Collapse
|
135
|
Cheng N, Watkins-Schulz R, Junkins RD, David CN, Johnson BM, Montgomery SA, Peine KJ, Darr DB, Yuan H, McKinnon KP, Liu Q, Miao L, Huang L, Bachelder EM, Ainslie KM, Ting JPY. A nanoparticle-incorporated STING activator enhances antitumor immunity in PD-L1-insensitive models of triple-negative breast cancer. JCI Insight 2018; 3:120638. [PMID: 30429378 DOI: 10.1172/jci.insight.120638] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 10/19/2018] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancer (TNBC) has few therapeutic options, and alternative approaches are urgently needed. Stimulator of IFN genes (STING) is becoming an exciting target for therapeutic adjuvants. However, STING resides inside the cell, and the intracellular delivery of CDNs, such as cGAMP, is required for the optimal activation of STING. We show that liposomal nanoparticle-delivered cGAMP (cGAMP-NP) activates STING more effectively than soluble cGAMP. These particles induce innate and adaptive host immune responses to preexisting tumors in both orthotopic and genetically engineered models of basal-like TNBC. cGAMP-NPs also reduce melanoma tumor load, with limited responsivity to anti-PD-L1. Within the tumor microenvironment, cGAMP-NPs direct both mouse and human macrophages (M), reprograming from protumorigenic M2-like phenotype toward M1-like phenotype; enhance MHC and costimulatory molecule expression; reduce M2 biomarkers; increase IFN-γ-producing T cells; augment tumor apoptosis; and increase CD4+ and CD8+ T cell infiltration. Activated T cells are required for tumor suppression, as their depletion reduces antitumor activity. Importantly, cGAMP-NPs prevent the formation of secondary tumors, and a single dose is sufficient to inhibit TNBC. These data suggest that a minimal system comprised of cGAMP-NP alone is sufficient to modulate the tumor microenvironment to effectively control PD-L1-insensitive TNBC.
Collapse
Affiliation(s)
- Ning Cheng
- Oral and Craniofacial Biomedicine Program, School of Dentistry.,Lineberger Comprehensive Cancer Center
| | | | | | | | | | | | - Kevin J Peine
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics
| | | | - Hong Yuan
- Department of Radiology.,Biomedical Imaging Research Center
| | | | - Qi Liu
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics.,University of North Carolina/North Carolina State University Joint Department of Biomedical Engineering
| | - Lei Miao
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics
| | - Leaf Huang
- Lineberger Comprehensive Cancer Center.,Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics
| | - Eric M Bachelder
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics
| | - Kristy M Ainslie
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics
| | - Jenny P-Y Ting
- Oral and Craniofacial Biomedicine Program, School of Dentistry.,Lineberger Comprehensive Cancer Center.,Curriculum of Genetics and Molecular Biology.,Department of Microbiology and Immunology.,Department of Genetics.,Institute for Inflammatory Diseases, and.,Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
136
|
Xu C, Li X, Liu P, Li M, Luo F. Patient-derived xenograft mouse models: A high fidelity tool for individualized medicine. Oncol Lett 2018; 17:3-10. [PMID: 30655732 PMCID: PMC6313209 DOI: 10.3892/ol.2018.9583] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 05/16/2017] [Indexed: 12/31/2022] Open
Abstract
Patient-derived xenograft (PDX) mouse models involve the direct transfer of fresh human tumor samples into immunodeficient mice following surgical resection or other medical operations. Gene expression in tumors may be maintained by serial passages of tumors from mouse to mouse. These models aid research into tumor biology and pharmacology without manual manipulation of cell cultures in vitro. and are widely used in individualized cancer therapy/translational medicine, drug development and coclinical trials. PDX models exhibit higher predictive values for clinical outcomes than cell line-derived xenograft models and genetically engineered mouse models. However, PDX models are associated with certain challenges in clinical application. The present study reviewed current collections of PDX models and assessed the challenges and future directions of this field.
Collapse
Affiliation(s)
- Cong Xu
- Department of Acute Abdomen Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Xuelu Li
- Department of Breast Surgery and Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Pixu Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,College of Pharmacy, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Man Li
- Department of Breast Surgery and Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Fuwen Luo
- Department of Acute Abdomen Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| |
Collapse
|
137
|
Grandori C, Kemp CJ. Personalized Cancer Models for Target Discovery and Precision Medicine. Trends Cancer 2018; 4:634-642. [PMID: 30149881 PMCID: PMC6242713 DOI: 10.1016/j.trecan.2018.07.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022]
Abstract
Although cancer research is progressing at an exponential rate, translating this knowledge to develop better cancer drugs and more effectively match drugs to patients is lagging. Genome profiling of tumors provides a snapshot of the genetic complexity of individual tumors, yet this knowledge is insufficient to guide therapy for most patients. Model systems, usually cancer cell lines or mice, have been instrumental in cancer research and drug development, but translation of results to the clinic is inefficient, in part, because these models do not sufficiently reflect the complexity and heterogeneity of human cancer. Here, we discuss the potential of combining genomics with high-throughput functional testing of patient-derived tumor cells to overcome key roadblocks in both drug target discovery and precision medicine.
Collapse
|
138
|
Tar PD, Thacker NA, Babur M, Watson Y, Cheung S, Little RA, Gieling RG, Williams KJ, O’Connor JPB. A new method for the high-precision assessment of tumor changes in response to treatment. Bioinformatics 2018; 34:2625-2633. [PMID: 29547950 PMCID: PMC6061877 DOI: 10.1093/bioinformatics/bty115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 02/05/2018] [Accepted: 03/12/2018] [Indexed: 11/12/2022] Open
Abstract
Motivation Imaging demonstrates that preclinical and human tumors are heterogeneous, i.e. a single tumor can exhibit multiple regions that behave differently during both development and also in response to treatment. The large variations observed in control group, tumors can obscure detection of significant therapeutic effects due to the ambiguity in attributing causes of change. This can hinder development of effective therapies due to limitations in experimental design rather than due to therapeutic failure. An improved method to model biological variation and heterogeneity in imaging signals is described. Specifically, linear Poisson modeling (LPM) evaluates changes in apparent diffusion co-efficient between baseline and 72 h after radiotherapy, in two xenograft models of colorectal cancer. The statistical significance of measured changes is compared to those attainable using a conventional t-test analysis on basic apparent diffusion co-efficient distribution parameters. Results When LPMs were applied to treated tumors, the LPMs detected highly significant changes. The analyses were significant for all tumors, equating to a gain in power of 4-fold (i.e. equivalent to having a sample size 16 times larger), compared with the conventional approach. In contrast, highly significant changes are only detected at a cohort level using t-tests, restricting their potential use within personalized medicine and increasing the number of animals required during testing. Furthermore, LPM enabled the relative volumes of responding and non-responding tissue to be estimated for each xenograft model. Leave-one-out analysis of the treated xenografts provided quality control and identified potential outliers, raising confidence in LPM data at clinically relevant sample sizes. Availability and implementation TINA Vision open source software is available from www.tina-vision.net. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- P D Tar
- Division of Informatics, Imaging and Data Science, Manchester Pharmacy School, Manchester, UK
| | - N A Thacker
- Division of Informatics, Imaging and Data Science, Manchester Pharmacy School, Manchester, UK
| | - M Babur
- Division of Pharmacy and Optometry, Manchester Pharmacy School, Manchester, UK
| | - Y Watson
- Division of Informatics, Imaging and Data Science, Manchester Pharmacy School, Manchester, UK
| | - S Cheung
- Division of Informatics, Imaging and Data Science, Manchester Pharmacy School, Manchester, UK
| | - R A Little
- Division of Informatics, Imaging and Data Science, Manchester Pharmacy School, Manchester, UK
| | - R G Gieling
- Division of Pharmacy and Optometry, Manchester Pharmacy School, Manchester, UK
| | - K J Williams
- Division of Pharmacy and Optometry, Manchester Pharmacy School, Manchester, UK
- Division of Cancer Sciences, University of Manchester
| | - J P B O’Connor
- Division of Cancer Sciences, University of Manchester
- Department of Radiology, The Christie Hospital NHS Trust, Manchester, UK
| |
Collapse
|
139
|
Best SA, Harapas CR, Kersbergen A, Rathi V, Asselin-Labat ML, Sutherland KD. FGFR3-TACC3 is an oncogenic fusion protein in respiratory epithelium. Oncogene 2018; 37:6096-6104. [PMID: 29991799 PMCID: PMC6215478 DOI: 10.1038/s41388-018-0399-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 05/24/2018] [Accepted: 06/08/2018] [Indexed: 11/25/2022]
Abstract
Structural rearrangements of the genome can drive lung tumorigenesis
through the generation of fusion genes with oncogenic properties. Advanced
genomic approaches have identified the presence of a genetic fusion between
fibroblast growth factor receptor 3
(FGFR3) and transforming acidic coiled-coil 3
(TACC3) in non-small cell lung cancer (NSCLC), providing a
novel target for FGFR inhibition. To interrogate the functional consequences of
the FGFR3-TACC3 fusion in the transformation of lung epithelial cells, we
generated a novel transgenic mouse model that expresses FGFR3-TACC3 concomitant
with loss of the p53 tumor suppressor gene. Intra-nasal
delivery of an Ad5-CMV-Cre virus promoted seromucinous glandular transformation
of olfactory cells lining the nasal cavities of FGFR3-TACC3
(LSL-F3T3) mice, which was further
accelerated upon loss of p53
(LSL-F3T3/p53).
Surprisingly, lung tumors failed to develop in intra-nasally infected
LSL-F3T3 and
LSL-F3T3/p53 mice. In
line with these observations, we demonstrated that intra-nasal delivery of
Ad5-CMV-Cre induces widespread Cre-mediated recombination in the olfactory
epithelium. Intra-tracheal delivery of Ad5-CMV-Cre into the lungs of
LSL-F3T3 and
LSL-F3T3/p53 mice
however, resulted in the development of lung adenocarcinomas. Taken together,
these findings provide in vivo evidence for an oncogenic
function of FGFR3-TACC3 in respiratory epithelium.
Collapse
Affiliation(s)
- Sarah A Best
- ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Cassandra R Harapas
- ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Ariena Kersbergen
- ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Vivek Rathi
- Department of Anatomical Pathology, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Marie-Liesse Asselin-Labat
- ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Kate D Sutherland
- ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia. .,Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
140
|
Ibarrola-Villava M, Cervantes A, Bardelli A. Preclinical models for precision oncology. Biochim Biophys Acta Rev Cancer 2018; 1870:239-246. [PMID: 29959990 DOI: 10.1016/j.bbcan.2018.06.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/17/2018] [Accepted: 06/18/2018] [Indexed: 12/15/2022]
Abstract
Precision medicine approaches have revolutionized oncology. Personalized treatments require not only identification of the driving molecular alterations, but also development of targeted therapies and diagnostic tests to identify the appropriate patient populations for clinical trials and subsequent therapeutic implementation. Preclinical in vitro and in vivo models are widely used to predict efficacy of newly developed treatments. Here we discuss whether, and to what extent, preclinical models including cell lines, organoids and tumorgrafts recapitulate key features of human tumors. The potential of preclinical models to anticipate treatment efficacy and clinical benefit is also presented, using examples in different tumor types.
Collapse
Affiliation(s)
- Maider Ibarrola-Villava
- Department of Oncology, Biomedical Research Institute - INCLIVA, University of Valencia, Valencia, Spain; Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy; centro de investigación biomedical en red CIBERONC, Spain.
| | - Andrés Cervantes
- Department of Oncology, Biomedical Research Institute - INCLIVA, University of Valencia, Valencia, Spain; centro de investigación biomedical en red CIBERONC, Spain
| | - Alberto Bardelli
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy; Department of Oncology, University of Torino, SP 142 km 3.95, Candiolo, TO, Italy.
| |
Collapse
|
141
|
Molecular phenotyping of laboratory mouse strains using 500 multiple reaction monitoring mass spectrometry plasma assays. Commun Biol 2018; 1:78. [PMID: 30271959 PMCID: PMC6123701 DOI: 10.1038/s42003-018-0087-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 05/28/2018] [Indexed: 12/18/2022] Open
Abstract
Mouse is the predominant experimental model for the study of human disease due, in part, to phylogenetic relationship, ease of breeding, and the availability of molecular tools for genetic manipulation. Advances in genome-editing methodologies, such as CRISPR-Cas9, enable the rapid production of new transgenic mouse strains, necessitating complementary high-throughput and systematic phenotyping technologies. In contrast to traditional protein phenotyping techniques, multiple reaction monitoring (MRM) mass spectrometry can be highly multiplexed without forgoing specificity or quantitative precision. Here we present MRM assays for the quantitation of 500 proteins and subsequently determine reference concentration values for plasma proteins across five laboratory mouse strains that are typically used in biomedical research, revealing inter-strain and intra-strain phenotypic differences. These 500 MRM assays will have a broad range of research applications including high-throughput phenotypic validation of novel transgenic mice, identification of candidate biomarkers, and general research applications requiring multiplexed and precise protein quantification.
Collapse
|
142
|
Yu C, Stefanson O, Liu Y, Wang ZA. Novel Method of Plasmid DNA Delivery to Mouse Bladder Urothelium by Electroporation. J Vis Exp 2018. [PMID: 29782018 DOI: 10.3791/57649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Genetically engineered mouse models (GEMMs) are extremely valuable in revealing novel biological insights into the initiation and progression mechanisms of human diseases such as cancer. Transgenic and conditional knockout mice have been frequently used for gene overexpression or ablation in specific tissues or cell types in vivo. However, generating germline mouse models can be time-consuming and costly. Recent advancements in gene editing technologies and the feasibility of delivering DNA plasmids by viral infection have enabled rapid generation of non-germline autochthonous mouse cancer models for several organs. The bladder is an organ that has been difficult for viral vectors to access, due to the presence of a glycosaminoglycan layer covering the urothelium. Here, we describe a novel method developed in lab for efficient delivery of DNA plasmids into the mouse bladder urothelium in vivo. Through intravesical instillation of pCAG-GFP DNA plasmid and electroporation of surgically exposed bladder, we show that the DNA plasmid can be delivered specifically into the bladder urothelial cells for transient expression. Our method provides a fast and convenient way for overexpression and knockdown of genes in the mouse bladder, and can be applied to building GEMMs of bladder cancer and other urological diseases.
Collapse
Affiliation(s)
- Chuan Yu
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz
| | - Ofir Stefanson
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz
| | - Yueli Liu
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz
| | - Zhu A Wang
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz;
| |
Collapse
|
143
|
Abstract
Cellular senescence is a highly stable cell cycle arrest that is elicited in response to different stresses. By imposing a growth arrest, senescence limits the replication of old or damaged cells. Besides exiting the cell cycle, senescent cells undergo many other phenotypic alterations such as metabolic reprogramming, chromatin rearrangement, or autophagy modulation. In addition, senescent cells produce and secrete a complex combination of factors, collectively referred as the senescence-associated secretory phenotype, that mediate most of their non-cell-autonomous effects. Because senescent cells influence the outcome of a variety of physiological and pathological processes, including cancer and age-related diseases, pro-senescent and anti-senescent therapies are actively being explored. In this Review, we discuss the mechanisms regulating different aspects of the senescence phenotype and their functional implications. This knowledge is essential to improve the identification and characterization of senescent cells in vivo and will help to develop rational strategies to modulate the senescence program for therapeutic benefit.
Collapse
Affiliation(s)
- Nicolás Herranz
- MRC London Institute of Medical Sciences, London, United Kingdom.,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Jesús Gil
- MRC London Institute of Medical Sciences, London, United Kingdom.,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
144
|
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
|
145
|
Sebolt-Leopold JS. Development of Preclinical Models to Understand and Treat Colorectal Cancer. Clin Colon Rectal Surg 2018; 31:199-204. [PMID: 29720906 DOI: 10.1055/s-0037-1602240] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The establishment and validation of preclinical models that faithfully recapitulate the pathogenesis and treatment response of human colorectal cancer (CRC) is critical to expedient therapeutic advances in the clinical management of this disease. Integral to the application of precision medicine for patients diagnosed with metastatic CRC is the need to understand the molecular determinants of response for a given therapy. Preclinical models of CRC have proven invaluable in answering many of our basic questions relating to the molecular aberrations that drive colorectal tumor progression. This review will address the comparative merits and limitations of the broad spectrum of in vitro and in vivo models available for study of colorectal tumors and their response to experimental therapies.
Collapse
Affiliation(s)
- Judith S Sebolt-Leopold
- Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
146
|
Soleimani S, Shamsi M, Ghazani MA, Modarres HP, Valente KP, Saghafian M, Ashani MM, Akbari M, Sanati-Nezhad A. Translational models of tumor angiogenesis: A nexus of in silico and in vitro models. Biotechnol Adv 2018; 36:880-893. [PMID: 29378235 DOI: 10.1016/j.biotechadv.2018.01.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 01/10/2018] [Accepted: 01/20/2018] [Indexed: 12/13/2022]
Abstract
Emerging evidence shows that endothelial cells are not only the building blocks of vascular networks that enable oxygen and nutrient delivery throughout a tissue but also serve as a rich resource of angiocrine factors. Endothelial cells play key roles in determining cancer progression and response to anti-cancer drugs. Furthermore, the endothelium-specific deposition of extracellular matrix is a key modulator of the availability of angiocrine factors to both stromal and cancer cells. Considering tumor vascular network as a decisive factor in cancer pathogenesis and treatment response, these networks need to be an inseparable component of cancer models. Both computational and in vitro experimental models have been extensively developed to model tumor-endothelium interactions. While informative, they have been developed in different communities and do not yet represent a comprehensive platform. In this review, we overview the necessity of incorporating vascular networks for both in vitro and in silico cancer models and discuss recent progresses and challenges of in vitro experimental microfluidic cancer vasculature-on-chip systems and their in silico counterparts. We further highlight how these two approaches can merge together with the aim of presenting a predictive combinatorial platform for studying cancer pathogenesis and testing the efficacy of single or multi-drug therapeutics for cancer treatment.
Collapse
Affiliation(s)
- Shirin Soleimani
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Milad Shamsi
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, AB T2N 1N4, Canada; Department of Mechanical Engineering, Isfahan University of Technology, Isfahan 8415683111, Iran
| | - Mehran Akbarpour Ghazani
- Department of Mechanical Engineering, Isfahan University of Technology, Isfahan 8415683111, Iran
| | - Hassan Pezeshgi Modarres
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Karolina Papera Valente
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Mohsen Saghafian
- Department of Mechanical Engineering, Isfahan University of Technology, Isfahan 8415683111, Iran
| | - Mehdi Mohammadi Ashani
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Amir Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, AB T2N 1N4, Canada.
| |
Collapse
|
147
|
Kawasaki K, Fujii M, Sato T. Gastroenteropancreatic neuroendocrine neoplasms: genes, therapies and models. Dis Model Mech 2018; 11:11/2/dmm029595. [PMID: 29590641 PMCID: PMC5894937 DOI: 10.1242/dmm.029595] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) refer to a group of heterogeneous cancers of neuroendocrine cell phenotype that mainly fall into one of two subtypes: gastroenteropancreatic neuroendocrine tumors (GEP-NETs; well differentiated) or gastroenteropancreatic neuroendocrine carcinomas (GEP-NECs; poorly differentiated). Although originally defined as orphan cancers, their steadily increasing incidence highlights the need to better understand their etiology. Accumulating epidemiological and clinical data have shed light on the pathological characteristics of these diseases. However, the relatively low number of patients has hampered conducting large-scale clinical trials and hence the development of novel treatment strategies. To overcome this limitation, tractable disease models that faithfully reflect clinical features of these diseases are needed. In this Review, we summarize the current understanding of the genetics and biology of these diseases based on conventional disease models, such as genetically engineered mouse models (GEMMs) and cell lines, and discuss the phenotypic differences between the models and affected humans. We also highlight the emerging disease models derived from human clinical samples, including patient-derived xenograft models and organoids, which may provide biological and therapeutic insights into GEP-NENs.
Collapse
Affiliation(s)
- Kenta Kawasaki
- Department of Gastroenterology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masayuki Fujii
- Department of Gastroenterology, Keio University School of Medicine, Tokyo 160-8582, Japan.,Department of Surgical Oncology, The University of Tokyo, Tokyo 113-8654, Japan
| | - Toshiro Sato
- Department of Gastroenterology, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
148
|
Yu ZH, Zhang ZY. Regulatory Mechanisms and Novel Therapeutic Targeting Strategies for Protein Tyrosine Phosphatases. Chem Rev 2018; 118:1069-1091. [PMID: 28541680 PMCID: PMC5812791 DOI: 10.1021/acs.chemrev.7b00105] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
An appropriate level of protein phosphorylation on tyrosine is essential for cells to react to extracellular stimuli and maintain cellular homeostasis. Faulty operation of signal pathways mediated by protein tyrosine phosphorylation causes numerous human diseases, which presents enormous opportunities for therapeutic intervention. While the importance of protein tyrosine kinases in orchestrating the tyrosine phosphorylation networks and in target-based drug discovery has long been recognized, the significance of protein tyrosine phosphatases (PTPs) in cellular signaling and disease biology has historically been underappreciated, due to a large extent to an erroneous assumption that they are largely constitutive and housekeeping enzymes. Here, we provide a comprehensive examination of a number of regulatory mechanisms, including redox modulation, allosteric regulation, and protein oligomerization, that control PTP activity. These regulatory mechanisms are integral to the myriad PTP-mediated biochemical events and reinforce the concept that PTPs are indispensable and specific modulators of cellular signaling. We also discuss how disruption of these PTP regulatory mechanisms can cause human diseases and how these diverse regulatory mechanisms can be exploited for novel therapeutic development.
Collapse
Affiliation(s)
- Zhi-Hong Yu
- Department of Medicinal Chemistry and Molecular Pharmacology, Department of Chemistry, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Department of Chemistry, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907
| |
Collapse
|
149
|
Patient-derived xenograft models of colorectal cancer in pre-clinical research: a systematic review. Oncotarget 2018; 7:66212-66225. [PMID: 27517155 PMCID: PMC5323228 DOI: 10.18632/oncotarget.11184] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 07/18/2016] [Indexed: 12/17/2022] Open
Abstract
AIMS We sought to objectively assess the internal and external validity of patient-derived xenograft (PDX) models as a platform in pre-clinical research into colorectal cancer (CRC). Metastatic disease is the most common cause of death from CRC, and despite significant research, the results of current combination chemotherapy and targeted therapies have been underwhelming for most of this patient group. One of the key factors limiting the success of translational CRC research is the biologically inaccurate models in which new therapies are developed. METHODS We used the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) checklist and SYRCLE (Systematic Review Centre for Laboratory animal Experimentation) guidelines to search Ovid MEDLINE and Embase databases up to July 2015 to identify studies involving PDX models of CRC where the model had been validated across multiple parameters. Data was extracted including host mouse strain, engraftment rate, site of engraftment, donor tumour source and development of metastases in the model. RESULTS Thirteen articles satisfied the inclusion criteria. There was significant heterogeneity amongst the included studies, but overall the median engraftment rate was high (70%) and PDX models faithfully recapitulated the characteristics of their patient tumours on the microscopic, genetic and functional levels. CONCLUSIONS PDX models of CRC have a reasonable internal validity and a high external validity. Developments in xenografting technology are broadening the applications of the PDX platform. However, the included studies could be improved by standardising reporting standards and closed following the ARRIVE (Animals in Research: Reporting In Vivo Experiments) guidelines.
Collapse
|
150
|
Jäger W, Moskalev I, Raven P, Goriki A, Bidnur S, Black PC. Orthotopic Mouse Models of Urothelial Cancer. Methods Mol Biol 2018; 1655:177-197. [PMID: 28889387 DOI: 10.1007/978-1-4939-7234-0_15] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Orthotopic mouse models of urothelial cancer are essential for testing novel therapies and molecular manipulations of cell lines in vivo. These models are either established by orthotopic inoculation of human (xenograft models) or murine tumor cells (syngeneic models) in immunocompromised or immune competent mice. Current techniques rely on inoculation by intravesical instillation or direct injection into the bladder wall. Alternative models include the induction of murine bladder tumors by chemical carcinogens (BBN) or genetic engineering (GEM).
Collapse
Affiliation(s)
- Wolfgang Jäger
- Department of Urology and Paediatric Urology, Johannes Gutenberg-Universität Mainz, Langenbeckstraße 1, 55131, Mainz, Germany.
| | - Igor Moskalev
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Peter Raven
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Akihiro Goriki
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Samir Bidnur
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Peter C Black
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|