101
|
Yan S, Yan J, Liu D, Li X, Kang Q, You W, Zhang J, Wang L, Tian Z, Lu W, Liu W, He W. A nano-predator of pathological MDMX construct by clearable supramolecular gold(I)-thiol-peptide complexes achieves safe and potent anti-tumor activity. Theranostics 2021; 11:6833-6846. [PMID: 34093856 PMCID: PMC8171083 DOI: 10.7150/thno.59020] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022] Open
Abstract
As alternatives to small-molecular proteolysis-targeting chimeras (PROTAC), peptide-based molecular glues (MG) are a broad range of dual-functional ligands that simultaneously bind with targetable proteins and E3 ligases by mimicking proteinprotein interaction (PPI) partners. Methods: Herein, we design a peptide-derived MG to target a tumor-driving protein, MDMX, for degradation, and nanoengineered it into a supramolecular gold(I)-thiol-peptide complex (Nano-MP) to implement the proteolysis recalcitrance, cellular internalization, and glutathione-triggered release. To optimize the tumor targeting, a pH-responsive macromolecule termed polyacryl sulfydryl imidazole (PSI) was synthesized to coat Nano-MP. Results: As expected, Nano-MP@PSI induced the MDMX degradation by ubiquitination and subsequently restored the anti-cancer function of p53 and p73. Nano-MP@PSI revealed potent anti-cancer activities in an orthotopic xenograft mouse model of retinoblastoma by intraocular injection and a patient-derived xenograft model of malignant pancreatic cancer by systemic injection, while maintaining a favorable safety profile and showing a highly favorable clearable profile of excretion from the living body. Conclusion: Collectively, this work not only provided a clinically viable paradigm for the treatment of a wide variety of tumors by multiple administration types, but, more importantly, it bridged the chasm between peptides and PROTACs, and likely reinvigorated the development of peptide-derived proteolysis-targeting chimeras for a great variety of diseases.
Collapse
Affiliation(s)
- Siqi Yan
- Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Ophthalmology Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin Yan
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Dan Liu
- Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiang Li
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Qianyan Kang
- Ophthalmology Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Weiming You
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Jinghua Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Lei Wang
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Zhiqi Tian
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, 45267 OH, USA
| | - Wuyuan Lu
- School of Basic Medicine, Fudan University, Shanghai 20433, China
| | - Wenjia Liu
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Wangxiao He
- Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
102
|
Chen D, Tan Y, Li Z, Li W, Yu L, Chen W, Liu Y, Liu L, Guo L, Huang W, Zhao Y. Organoid Cultures Derived From Patients With Papillary Thyroid Cancer. J Clin Endocrinol Metab 2021; 106:1410-1426. [PMID: 33524147 DOI: 10.1210/clinem/dgab020] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Indexed: 02/08/2023]
Abstract
CONTEXT Papillary thyroid cancer (PTC) has been one of the most frequent endocrine malignancies around the world. Although most PTC patients have a favorable prognosis, a subgroup of patients die, especially when disease recurrence occurs. There is a pressing need for clinically relevant preclinical thyroid cancer models for personalized therapy because of the lack of in vitro models that faithfully represent the biology of the parental tumors. OBJECTIVE To understand thyroid cancer and translate this knowledge to clinical applications, patient-derived PTC organoids as a promising new preclinical model were established. METHODS Surgically resected PTC primary tissues were dissociated and processed for organoid derivation. Tumor organoids were subsequently subjected to histological characterization, DNA sequencing, drug screen, and cell proliferation assay, respectively. RESULTS We describe a 3-dimensional culture system for the long-term expansion of patient-derived PTC organoid lines. Notably, PTC organoids preserve the histopathological profiles and genomic heterogeneity of the originating tumors. Drug sensitivity assays of PTC organoids demonstrate patient-specific drug responses, and large correlations with the respective mutational profiles. Estradiol was shown to promote cell proliferation of PTC organoids in the presence of estrogen receptor α (ERα), regardless of the expression of ERβ and G protein-coupled ER. CONCLUSION These data suggest that these newly developed PTC-derived organoids may be an excellent preclinical model for studying clinical response to anticancer drugs in a personalized way, as well as provide a potential strategy to develop prevention and treatment options for thyroid cancer with ERα-specific antagonists.
Collapse
Affiliation(s)
- Dong Chen
- Institute of Shenzhen Translational Medicine, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Yawen Tan
- Department of Breast and Thyroid Surgery, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Zhichao Li
- Institute of Shenzhen Translational Medicine, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Wujiao Li
- Institute of Shenzhen Translational Medicine, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Lei Yu
- Institute of Shenzhen Translational Medicine, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Wei Chen
- Institute of Shenzhen Translational Medicine, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Yuchen Liu
- Institute of Shenzhen Translational Medicine, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Lisa Liu
- Institute of Shenzhen Translational Medicine, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Liangfeng Guo
- Department of Breast and Thyroid Surgery, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Weiren Huang
- Institute of Shenzhen Translational Medicine, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Yongsheng Zhao
- Institute of Shenzhen Translational Medicine, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
103
|
Lei X, Ou Z, Yang Z, Zhong J, Zhu Y, Tian J, Wu J, Deng H, Lin X, Peng Y, Li B, He L, Tu Z, Chen W, Li Q, Liu N, Zhang H, Wang Z, Fang Z, Yamada T, Lv X, Tian T, Pan G, Wu F, Xiao L, Zhang L, Cai T, Wang X, Tannous BA, Li J, Kontos F, Ferrone S, Fan S. A Pan-Histone Deacetylase Inhibitor Enhances the Antitumor Activity of B7-H3-Specific CAR T Cells in Solid Tumors. Clin Cancer Res 2021; 27:3757-3771. [PMID: 33811153 DOI: 10.1158/1078-0432.ccr-20-2487] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/03/2020] [Accepted: 03/29/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE The limited efficacy of chimeric antigen receptor (CAR) T-cell therapies with solid malignancies prompted us to test whether epigenetic therapy could enhance the antitumor activity of B7-H3.CAR T cells with several solid cancer types. EXPERIMENTAL DESIGN We evaluated B7-H3 expression in many human solid cancer and normal tissue samples. The efficacy of the combinatorial therapy with B7-H3.CAR T cells and the deacetylase inhibitor SAHA with several solid cancer types and the potential underlying mechanisms were characterized with in vitro and ex vivo experiments. RESULTS B7-H3 is expressed in most of the human solid tumor samples tested, but exhibits a restricted expression in normal tissues. B7-H3.CAR T cells selectively killed B7-H3 expressing human cancer cell lines in vitro. A low dose of SAHA upregulated B7-H3 expression in several types of solid cancer cells at the transcriptional level and B7-H3.CAR expression on human transgenic T-cell membrane. In contrast, the expression of immunosuppressive molecules, such as CTLA-4 and TET2, by T cells was downregulated upon SAHA treatment. A low dose of SAHA significantly enhanced the antitumor activity of B7-H3.CAR T cells with solid cancers in vitro and ex vivo, including orthotopic patient-derived xenograft and metastatic models treated with autologous CAR T-cell infusions. CONCLUSIONS Our results show that our novel strategy which combines SAHA and B7-H3.CAR T cells enhances their therapeutic efficacy with solid cancers and justify its translation to a clinical setting.
Collapse
Affiliation(s)
- Xinyuan Lei
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China.,State University of New York at Stony Brook, Stony Brook, New York
| | - Zhanpeng Ou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Zhaohui Yang
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianglong Zhong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Yanliang Zhu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Jing Tian
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiannan Wu
- Department of Breast Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Heran Deng
- Department of Breast Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xinyu Lin
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Peng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Bowen Li
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lile He
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Zhiming Tu
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Weixiong Chen
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qunxing Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Niu Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Hanqing Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Zhangsong Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Zezhen Fang
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Teppei Yamada
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Xiaobin Lv
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, Center Laboratory, the Third Affiliated Hospital, Nanchang University, Nanchang, China
| | - Tian Tian
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guokai Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Fan Wu
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liping Xiao
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lizao Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Tingting Cai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Xinhui Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bakhos A Tannous
- Experimental Therapeutics and Molecular Imaging Lab, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jinsong Li
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Filippos Kontos
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Song Fan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China. .,Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
104
|
Takahashi N, Higa A, Hiyama G, Tamura H, Hoshi H, Dobashi Y, Katahira K, Ishihara H, Takagi K, Goda K, Okabe N, Muto S, Suzuki H, Shimomura K, Watanabe S, Takagi M. Construction of in vitro patient-derived tumor models to evaluate anticancer agents and cancer immunotherapy. Oncol Lett 2021; 21:406. [PMID: 33841567 PMCID: PMC8020396 DOI: 10.3892/ol.2021.12667] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
An in vitro assay system using patient-derived tumor models represents a promising preclinical cancer model that replicates the disease better than traditional cell culture models. Patient-derived tumor organoid (PDO) and patient-derived tumor xenograft (PDX) models have been previously established from different types of human tumors to recapitulate accurately and efficiently their tissue architecture and function. However, these models have low throughput and are challenging to construct. Thus, the present study aimed to establish a simple in vitro high-throughput assay system using PDO and PDX models. Furthermore, the current study aimed to evaluate different classes of anticancer drugs, including chemotherapeutic, molecular targeted and antibody drugs, using PDO and PDX models. First, an in vitro high-throughput assay system was constructed using PDO and PDX established from solid and hematopoietic tumors cultured in 384-well plates to evaluate anticancer agents. In addition, an in vitro evaluation system of the immune response was developed using PDO and PDX. Novel cancer immunotherapeutic agents with marked efficacy have been used against various types of tumor. Thus, there is an urgent need for in vitro functional potency assays that can simulate the complex interaction of immune cells with tumor cells and can rapidly test the efficacy of different immunotherapies or antibody drugs. An evaluation system for the antibody-dependent cellular cytotoxic activity of anti-epidermal growth factor receptor antibody and the cytotoxic activity of activated lymphocytes, such as cytotoxic T lymphocytes and natural killer cells, was constructed. Moreover, immune response assay systems with bispecific T-cell engagers were developed using effector cells. The present results demonstrated that in vitro assay systems using PDO and PDX may be suitable for evaluating anticancer agents and immunotherapy potency with high reproducibility and simplicity.
Collapse
Affiliation(s)
- Nobuhiko Takahashi
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima, Fukushima 960-1295, Japan.,Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University, Fukushima, Fukushima 960-1295, Japan
| | - Arisa Higa
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima, Fukushima 960-1295, Japan
| | - Gen Hiyama
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima, Fukushima 960-1295, Japan
| | - Hirosumi Tamura
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima, Fukushima 960-1295, Japan
| | - Hirotaka Hoshi
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima, Fukushima 960-1295, Japan
| | - Yuu Dobashi
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima, Fukushima 960-1295, Japan
| | - Kiyoaki Katahira
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima, Fukushima 960-1295, Japan
| | - Hiroya Ishihara
- Research and Development, Department of Biological Evaluation Technology 2, Olympus Corporation, Hachioji, Tokyo 192-8512, Japan
| | - Kosuke Takagi
- Research and Development, Department of Technology Innovation 3, Olympus Corporation, Hachioji, Tokyo 192-8512, Japan
| | - Kazuhito Goda
- Research and Development, Department of Biological Evaluation Technology 2, Olympus Corporation, Hachioji, Tokyo 192-8512, Japan
| | - Naoyuki Okabe
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima, Fukushima 960-1295, Japan
| | - Satoshi Muto
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima, Fukushima 960-1295, Japan
| | - Hiroyuki Suzuki
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima, Fukushima 960-1295, Japan
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University, Fukushima, Fukushima 960-1295, Japan
| | - Shinya Watanabe
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima, Fukushima 960-1295, Japan
| | - Motoki Takagi
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima, Fukushima 960-1295, Japan
| |
Collapse
|
105
|
de Souza JC, Miguita L, Gomez RS, Gomes CC. Patient-derived xenograft models for the study of benign human neoplasms. Exp Mol Pathol 2021; 120:104630. [PMID: 33744281 DOI: 10.1016/j.yexmp.2021.104630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/07/2021] [Accepted: 03/14/2021] [Indexed: 12/27/2022]
Abstract
Preclinical models are a core feature of translational research, and patient-derived xenograft (PDX) models have increasingly been used with such purpose. PDX involves the transplantation of fresh human tumor samples into immunodeficient mice to overcome immunologic rejection. It is a valuable tool for basic as well as preclinical research, contributing to the establishment of models to characterize the neoplasms to drug screening and to allow the identification of therapeutic targets. The use of these models is justified because they retain the histological and genomic features of the primary tumor. PDX models are well described for malignant neoplasms, for which the advantages are clear and include the development of drug treatments. The establishment of malignant tumors PDX is undeniably important from a medical perspective. However, few studies have used such models for benign neoplasms. The use of PDX for benign neoplasm studies can help to clarify the pathobiology of these diseases, as well as invasion and malignant transformation mechanisms, which from a biological perspective is equally important to the study of malignant tumors. Therefore, the aim of this study is to review the current methodology for PDX model generation and to cover its main applications, focusing on benign neoplasms.
Collapse
Affiliation(s)
- Juliana Cristina de Souza
- Department of Pathology, Biological Science Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.
| | - Lucyene Miguita
- Department of Pathology, Biological Science Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.
| | - Ricardo Santiago Gomez
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil..
| | - Carolina Cavaliéri Gomes
- Department of Pathology, Biological Science Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.
| |
Collapse
|
106
|
Investigating T Cell Immunity in Cancer: Achievements and Prospects. Int J Mol Sci 2021; 22:ijms22062907. [PMID: 33809369 PMCID: PMC7999898 DOI: 10.3390/ijms22062907] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/04/2021] [Accepted: 03/10/2021] [Indexed: 12/21/2022] Open
Abstract
T cells play a key role in tumour surveillance, both identifying and eliminating transformed cells. However, as tumours become established they form their own suppressive microenvironments capable of shutting down T cell function, and allowing tumours to persist and grow. To further understand the tumour microenvironment, including the interplay between different immune cells and their role in anti-tumour immune responses, a number of studies from mouse models to clinical trials have been performed. In this review, we examine mechanisms utilized by tumour cells to reduce their visibility to CD8+ Cytotoxic T lymphocytes (CTL), as well as therapeutic strategies trialled to overcome these tumour-evasion mechanisms. Next, we summarize recent advances in approaches to enhance CAR T cell activity and persistence over the past 10 years, including bispecific CAR T cell design and early evidence of efficacy. Lastly, we examine mechanisms of T cell infiltration and tumour regression, and discuss the strengths and weaknesses of different strategies to investigate T cell function in murine tumour models.
Collapse
|
107
|
Validation of a multicellular tumor microenvironment system for modeling patient tumor biology and drug response. Sci Rep 2021; 11:5535. [PMID: 33692370 PMCID: PMC7946945 DOI: 10.1038/s41598-021-84612-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
Lung cancer rates are rising globally and non-small cell lung cancer (NSCLC) has a five year survival rate of only 24%. Unfortunately, the development of drugs to treat cancer is severely hampered by the inefficiency of translating pre-clinical studies into clinical benefit. Thus, we sought to apply a tumor microenvironment system (TMES) to NSCLC. Using microvascular endothelial cells, lung cancer derived fibroblasts, and NSCLC tumor cells in the presence of in vivo tumor-derived hemodynamic flow and transport, we demonstrate that the TMES generates an in-vivo like biological state and predicts drug response to EGFR inhibitors. Transcriptomic and proteomic profiling indicate that the TMES recapitulates the in vivo and patient molecular biological state providing a mechanistic rationale for the predictive nature of the TMES. This work further validates the TMES for modeling patient tumor biology and drug response indicating utility of the TMES as a predictive tool for drug discovery and development and potential for use as a system for patient avatars.
Collapse
|
108
|
Lopes MB, Martins EP, Vinga S, Costa BM. The Role of Network Science in Glioblastoma. Cancers (Basel) 2021; 13:1045. [PMID: 33801334 PMCID: PMC7958335 DOI: 10.3390/cancers13051045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Network science has long been recognized as a well-established discipline across many biological domains. In the particular case of cancer genomics, network discovery is challenged by the multitude of available high-dimensional heterogeneous views of data. Glioblastoma (GBM) is an example of such a complex and heterogeneous disease that can be tackled by network science. Identifying the architecture of molecular GBM networks is essential to understanding the information flow and better informing drug development and pre-clinical studies. Here, we review network-based strategies that have been used in the study of GBM, along with the available software implementations for reproducibility and further testing on newly coming datasets. Promising results have been obtained from both bulk and single-cell GBM data, placing network discovery at the forefront of developing a molecularly-informed-based personalized medicine.
Collapse
Affiliation(s)
- Marta B. Lopes
- Center for Mathematics and Applications (CMA), FCT, UNL, 2829-516 Caparica, Portugal
- NOVA Laboratory for Computer Science and Informatics (NOVA LINCS), FCT, UNL, 2829-516 Caparica, Portugal
| | - Eduarda P. Martins
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (E.P.M.); (B.M.C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
| | - Susana Vinga
- INESC-ID, Instituto Superior Técnico, Universidade de Lisboa, 1000-029 Lisbon, Portugal;
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisbon, Portugal
| | - Bruno M. Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (E.P.M.); (B.M.C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
| |
Collapse
|
109
|
Chen X, Li Y, Yao T, Jia R. Benefits of Zebrafish Xenograft Models in Cancer Research. Front Cell Dev Biol 2021; 9:616551. [PMID: 33644052 PMCID: PMC7905065 DOI: 10.3389/fcell.2021.616551] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
As a promising in vivo tool for cancer research, zebrafish have been widely applied in various tumor studies. The zebrafish xenograft model is a low-cost, high-throughput tool for cancer research that can be established quickly and requires only a small sample size, which makes it favorite among researchers. Zebrafish patient-derived xenograft (zPDX) models provide promising evidence for short-term clinical treatment. In this review, we discuss the characteristics and advantages of zebrafish, such as their transparent and translucent features, the use of vascular fluorescence imaging, the establishment of metastatic and intracranial orthotopic models, individual pharmacokinetics measurements, and tumor microenvironment. Furthermore, we introduce how these characteristics and advantages are applied other in tumor studies. Finally, we discuss the future direction of the use of zebrafish in tumor studies and provide new ideas for the application of it.
Collapse
Affiliation(s)
- Xingyu Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yongyun Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Tengteng Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
110
|
Establishment and Long-Term Expansion of Small Cell Lung Cancer Patient-Derived Tumor Organoids. Int J Mol Sci 2021; 22:ijms22031349. [PMID: 33572899 PMCID: PMC7866263 DOI: 10.3390/ijms22031349] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
Differential chemo-sensitivity of cancer cells, which is attributed to the cellular heterogeneity and phenotypic variation of cancer cells, is considered to be the main reason for tumor recurrence after chemotherapy. Here, we generated small cell lung cancer patient-derived tumor organoids and subjected them to long-term expansion with the addition of WNT3A or R-spondin1. We confirmed that the organoids have similar genetic profiles, molecular characteristics, and morphological architectures to the corresponding patient tumor tissue during and after long-term expansion. Interestingly, the cellular heterogeneity of organoids is reflected in their differential response to cisplatin or etoposide. We propose to utilize the organoids as small cell lung cancer patient avatar models that would be ideal for investigating the mechanisms underlying tumor recurrence after chemotherapy, and would ultimately help to develop personalized medicine.
Collapse
|
111
|
Thai VL, Griffin KH, Thorpe SW, Randall RL, Leach JK. Tissue engineered platforms for studying primary and metastatic neoplasm behavior in bone. J Biomech 2021; 115:110189. [PMID: 33385867 PMCID: PMC7855491 DOI: 10.1016/j.jbiomech.2020.110189] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/02/2020] [Accepted: 12/11/2020] [Indexed: 12/19/2022]
Abstract
Cancer is the second leading cause of death in the United States, claiming more than 560,000 lives each year. Osteosarcoma (OS) is the most common primary malignant tumor of bone in children and young adults, while bone is a common site of metastasis for tumors initiating from other tissues. The heterogeneity, continual evolution, and complexity of this disease at different stages of tumor progression drives a critical need for physiologically relevant models that capture the dynamic cancer microenvironment and advance chemotherapy techniques. Monolayer cultures have been favored for cell-based research for decades due to their simplicity and scalability. However, the nature of these models makes it impossible to fully describe the biomechanical and biochemical cues present in 3-dimensional (3D) microenvironments, such as ECM stiffness, degradability, surface topography, and adhesivity. Biomaterials have emerged as valuable tools to model the behavior of various cancers by creating highly tunable 3D systems for studying neoplasm behavior, screening chemotherapeutic drugs, and developing novel treatment delivery techniques. This review highlights the recent application of biomaterials toward the development of tumor models, details methods for their tunability, and discusses the clinical and therapeutic applications of these systems.
Collapse
Affiliation(s)
- Victoria L Thai
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, United States
| | - Katherine H Griffin
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, United States; School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, United States
| | - Steven W Thorpe
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, United States
| | - R Lor Randall
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, United States
| | - J Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, United States; Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, United States.
| |
Collapse
|
112
|
Janke LJ, Imai DM, Tillman H, Doty R, Hoenerhoff MJ, Xu JJ, Freeman Z, Allen P, Fowlkes NW, Iacobucci I, Dickerson K, Mullighan CG, Vogel P, Rehg JE. Development of Mast Cell and Eosinophil Hyperplasia and HLH/MAS-Like Disease in NSG-SGM3 Mice Receiving Human CD34+ Hematopoietic Stem Cells or Patient-Derived Leukemia Xenografts. Vet Pathol 2021; 58:181-204. [PMID: 33208054 PMCID: PMC8414369 DOI: 10.1177/0300985820970144] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Immunocompromised mouse strains expressing human transgenes are being increasingly used in biomedical research. The genetic modifications in these mice cause various cellular responses, resulting in histologic features unique to each strain. The NSG-SGM3 mouse strain is similar to the commonly used NSG (NOD scid gamma) strain but expresses human transgenes encoding stem cell factor (also known as KIT ligand), granulocyte-macrophage colony-stimulating factor, and interleukin 3. This report describes 3 histopathologic features seen in these mice when they are unmanipulated or after transplantation with human CD34+ hematopoietic stem cells (HSCs), virally transduced hCD34+ HSCs, or a leukemia patient-derived xenograft. The first feature is mast cell hyperplasia: unmanipulated, naïve mice develop periductular pancreatic aggregates of murine mast cells, whereas mice given the aforementioned human cells develop a proliferative infiltrative interstitial pancreatic mast cell hyperplasia but with human mast cells. The second feature is the predisposition of NSG-SGM3 mice given these human cells to develop eosinophil hyperplasia. The third feature, secondary hemophagocytic lymphohistiocytosis/macrophage activation syndrome (HLH/MAS)-like disease, is the most pronounced in both its clinical and histopathologic presentations. As part of this disease, a small number of mice also have histiocytic infiltration of the brain and spinal cord with subsequent neurologic or vestibular signs. The presence of any of these features can confound accurate histopathologic interpretation; therefore, it is important to recognize them as strain characteristics and to differentiate them from what may be experimentally induced in the model being studied.
Collapse
Affiliation(s)
- Laura J. Janke
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Denise M. Imai
- Comparative Pathology Laboratory, University of California, Davis, California, USA
| | - Heather Tillman
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | | | - Mark J. Hoenerhoff
- In Vivo Animal Core, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jiajie J. Xu
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Zach Freeman
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Portia Allen
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Natalie Wall Fowlkes
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ilaria Iacobucci
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Kirsten Dickerson
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Charles G. Mullighan
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Hematological Malignancies Program, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Peter Vogel
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jerold E. Rehg
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
113
|
Manso J, Sharifi-Rad J, Zam W, Tsouh Fokou PV, Martorell M, Pezzani R. Plant Natural Compounds in the Treatment of Adrenocortical Tumors. Int J Endocrinol 2021; 2021:5516285. [PMID: 34567112 PMCID: PMC8463247 DOI: 10.1155/2021/5516285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 05/14/2021] [Accepted: 08/31/2021] [Indexed: 01/08/2023] Open
Abstract
Plant natural products are a plethora of diverse and complex molecules produced by the plant secondary metabolism. Among these, many can reserve beneficial or curative properties when employed to treat human diseases. Even in cancer, they can be successfully used and indeed numerous phytochemicals exert antineoplastic activity. The most common molecules derived from plants and used in the fight against cancer are polyphenols, i.e., quercetin, genistein, resveratrol, curcumin, etc. Despite valuable data especially in preclinical models on such compounds, few of them are currently used in the medical practice. Also, in adrenocortical tumors (ACT), phytochemicals are scarcely or not at all used. This work summarizes the available research on phytochemicals used against ACT and adrenocortical cancer, a very rare disease with poor prognosis and high metastatic potential, and wants to contribute to stimulate preclinical and clinical research to find new therapeutic strategies among the overabundance of biomolecules produced by the plant kingdom.
Collapse
Affiliation(s)
- Jacopo Manso
- Endocrinology Unit, Department of Medicine (DIMED), University of Padova, Via Ospedale 105, Padova 35128, Italy
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Facultad de Medicina, Universidad del Azuay, Cuenca, Ecuador
| | - Wissam Zam
- Analytical and Food Chemistry Department, Faculty of Pharmacy, Tartous University, Tartous, Syria
| | | | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, Centre for Healthy Living, University of Concepción, Concepción 4070386, Chile
- Universidad de Concepción, Unidad de Desarrollo Tecnológico, UDT, Concepción 4070386, Chile
| | - Raffaele Pezzani
- Endocrinology Unit, Department of Medicine (DIMED), University of Padova, Via Ospedale 105, Padova 35128, Italy
- Phytotherapy Lab, Endocrinology Unit, Department of Medicine (DIMED), University of Padova, via Ospedale 105, 35128 Padova, Italy
| |
Collapse
|
114
|
Forde S, Matthews JD, Jahangiri L, Lee LC, Prokoph N, Malcolm TIM, Giger OT, Bell N, Blair H, O'Marcaigh A, Smith O, Kenner L, Bomken S, Burke GAA, Turner SD. Paediatric Burkitt lymphoma patient-derived xenografts capture disease characteristics over time and are a model for therapy. Br J Haematol 2021; 192:354-365. [PMID: 32880915 DOI: 10.1111/bjh.17043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/26/2020] [Indexed: 12/28/2022]
Abstract
Burkitt lymphoma (BL) accounts for almost two-thirds of all B-cell non-Hodgkin lymphoma (B-NHL) in children and adolescents and is characterised by a MYC translocation and rapid cell turnover. Intensive chemotherapeutic regimens have been developed in recent decades, including the lymphomes malins B (LMB) protocol, which have resulted in a survival rate in excess of 90%. Recent clinical trials have focused on immunochemotherapy, with the addition of rituximab to chemotherapeutic backbones, showing encouraging results. Despite these advances, relapse and refractory disease occurs in up to 10% of patients and salvage options for these carry a dismal prognosis. Efforts to better understand the molecular and functional characteristics driving relapse and refractory disease may help improve this prognosis. This study has established a paediatric BL patient-derived xenograft (PDX) resource which captures and maintains tumour heterogeneity, may be used to better characterise tumours and identify cell populations responsible for therapy resistance.
Collapse
Affiliation(s)
- Sorcha Forde
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Jamie D Matthews
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Leila Jahangiri
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK.,Department of Life Sciences, Birmingham City University, Birmingham, UK
| | - Liam C Lee
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Nina Prokoph
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Tim I M Malcolm
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Olivier T Giger
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Natalie Bell
- Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Helen Blair
- Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Owen Smith
- Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Lukas Kenner
- Department of Pathology, Medical University of Vienna, Vienna, Austria.,Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria.,Christian Doppler Laboratory for Applied Metabolomics, Vienna, Austria
| | - Simon Bomken
- Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.,The Great North Children's Hospital, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.,Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Gladstone A A Burke
- Department of Paediatric Oncology and Haematology, Addenbrooke's Hospital, Cambridge, UK
| | - Suzanne D Turner
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK.,Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| |
Collapse
|
115
|
Han W, El Botty R, Montaudon E, Malaquin L, Deschaseaux F, Espagnolle N, Marangoni E, Cottu P, Zalcman G, Parrini MC, Assayag F, Sensebe L, Silberzan P, Vincent-Salomon A, Dutertre G, Roman-Roman S, Descroix S, Camonis J. In vitro bone metastasis dwelling in a 3D bioengineered niche. Biomaterials 2020; 269:120624. [PMID: 33421710 DOI: 10.1016/j.biomaterials.2020.120624] [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: 08/04/2020] [Revised: 12/06/2020] [Accepted: 12/18/2020] [Indexed: 12/28/2022]
Abstract
Bone is the most frequent metastasis site for breast cancer. As well as dramatically increasing disease burden, bone metastases are also an indicator of poor prognosis. One of the main challenges in investigating bone metastasis in breast cancer is engineering in vitro models that replicate the features of in vivo bone environments. Such in vitro models ideally enable the biology of the metastatic cells to mimic their in vivo behavior as closely as possible. Here, taking benefit of cutting-edge technologies both in microfabrication and cancer cell biology, we have developed an in vitro breast cancer bone-metastasis model. To do so we first 3D printed a bone scaffold that reproduces the trabecular architecture and that can be conditioned with osteoblast-like cells, a collagen matrix, and mineralized calcium. We thus demonstrated that this device offers an adequate soil to seed primary breast cancer bone metastatic cells. In particular, patient-derived xenografts being considered as a better approach than cell lines to achieve clinically relevant results, we demonstrate the ability of this biomimetic bone niche model to host patient-derived xenografted metastatic breast cancer cells. These patient-derived xenograft cells show a long-term survival in the bone model and maintain their cycling propensity, and exhibit the same modulated drug response as in vivo. This experimental system enables access to the idiosyncratic features of the bone microenvironment and cancer bone metastasis, which has implications for drug testing.
Collapse
Affiliation(s)
- Weijing Han
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005, Paris, France; ART Group, Inserm U830, 75005, Paris, France; Laboratoire PhysicoChimie Curie, Institut Curie, PSL Research University - Sorbonne Université - CNRS. Equipe Labellisée Ligue Contre le Cancer; 75005, Paris, France
| | - Rania El Botty
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, 75005, Paris, France
| | - Elodie Montaudon
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, 75005, Paris, France
| | - Laurent Malaquin
- LAAS-CNRS, Université de Toulouse, CNRS, F-31400, Toulouse, France
| | - Frederic Deschaseaux
- STROMALab, Etablissement Français Du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, ERL5311 CNRS, National Veterinary School of Toulouse (ENVT), Toulouse, France
| | - Nicolas Espagnolle
- STROMALab, Etablissement Français Du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, ERL5311 CNRS, National Veterinary School of Toulouse (ENVT), Toulouse, France
| | - Elisabetta Marangoni
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, 75005, Paris, France
| | - Paul Cottu
- Department of Medical Oncology, Institut Curie and Paris Sciences et Lettres Research University, 75005, Paris, France
| | - Gérard Zalcman
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005, Paris, France; ART Group, Inserm U830, 75005, Paris, France; Thoracic Oncology Department and Early Phase Unit CIC-1425, Hôpital Bichat, AP-HP, Université de Paris, 75018, Paris, France
| | - Maria Carla Parrini
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005, Paris, France; ART Group, Inserm U830, 75005, Paris, France
| | - Franck Assayag
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, 75005, Paris, France
| | - Luc Sensebe
- STROMALab, Etablissement Français Du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, ERL5311 CNRS, National Veterinary School of Toulouse (ENVT), Toulouse, France
| | - Pascal Silberzan
- Laboratoire PhysicoChimie Curie, Institut Curie, PSL Research University - Sorbonne Université - CNRS. Equipe Labellisée Ligue Contre le Cancer; 75005, Paris, France
| | - Anne Vincent-Salomon
- Department of Pathology, Institut Curie Hospital, 26, Rue D'Ulm, F-75248, Paris, France
| | - Guillaume Dutertre
- Surgical Oncology Department, Institut Curie, PSL Research University, 75005, Paris, France
| | - Sergio Roman-Roman
- Translational Research Department, Institut Curie, PSL Research University, 75005, Paris, France
| | - Stephanie Descroix
- Laboratoire PhysicoChimie Curie, Institut Curie, PSL Research University - Sorbonne Université - CNRS. Equipe Labellisée Ligue Contre le Cancer; 75005, Paris, France.
| | - Jacques Camonis
- Institut Curie, Centre de Recherche, Paris Sciences et Lettres Research University, 75005, Paris, France; ART Group, Inserm U830, 75005, Paris, France.
| |
Collapse
|
116
|
Bender A, Cortés-Ciriano I. Artificial intelligence in drug discovery: what is realistic, what are illusions? Part 1: Ways to make an impact, and why we are not there yet. Drug Discov Today 2020; 26:511-524. [PMID: 33346134 DOI: 10.1016/j.drudis.2020.12.009] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/07/2020] [Accepted: 12/11/2020] [Indexed: 12/30/2022]
Abstract
Although artificial intelligence (AI) has had a profound impact on areas such as image recognition, comparable advances in drug discovery are rare. This article quantifies the stages of drug discovery in which improvements in the time taken, success rate or affordability will have the most profound overall impact on bringing new drugs to market. Changes in clinical success rates will have the most profound impact on improving success in drug discovery; in other words, the quality of decisions regarding which compound to take forward (and how to conduct clinical trials) are more important than speed or cost. Although current advances in AI focus on how to make a given compound, the question of which compound to make, using clinical efficacy and safety-related end points, has received significantly less attention. As a consequence, current proxy measures and available data cannot fully utilize the potential of AI in drug discovery, in particular when it comes to drug efficacy and safety in vivo. Thus, addressing the questions of which data to generate and which end points to model will be key to improving clinically relevant decision-making in the future.
Collapse
Affiliation(s)
- Andreas Bender
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road CB2 1EW, UK; Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
| | - Isidro Cortés-Ciriano
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge CB10 1SD, UK.
| |
Collapse
|
117
|
Khalighfard S, Alizadeh AM, Poorkhani A, Motahari M, Tahmasebifar A, Omranipour R, Keshavarz P, Haddad P. Evaluation of the treatment strategies on patient-derived xenograft mice of human breast tumor. Eur J Pharmacol 2020; 889:173605. [PMID: 32980347 DOI: 10.1016/j.ejphar.2020.173605] [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: 04/14/2020] [Revised: 09/19/2020] [Accepted: 09/23/2020] [Indexed: 01/14/2023]
Abstract
Since only a minority of patients may respond to single-agent therapies, methods to test the potential antitumor activity of rational combination therapies are still needed. This study aimed to characterize the efficacy of antitumor combination therapies in vivo within the primary tumor using patient-derived xenograft (PDX) models by gamma-irradiation-induced immune suppression. We employed four Luminal A PDX models obtained from human mammary tumors grown in mice. PDX models were implanted into the right flank of mice, and treatments have ensued once tumor volume reached ~150 mm3. Four of the active drugs- Adriamycin, Cyclophosphamide, Taxotere, and Tamoxifen-were tested in vivo to treat mammary tumors. The tumor volume was measured during the study. The mice's immune system was inherently suppressed by gamma irradiation, thus allowing human tumors to grow. The results showed that the tumorigenesis rate of the PDX model was from 65 to 80%. PDX models were successfully established with a high frequency of tumor engraftment. Humanized mice treated with a two-drug regimen, that is, adriamycin + cyclophosphamide exhibited an increased antitumor response than a three-drug regimen, that is, adriamycin + cyclophosphamide + taxotere that correlated with tumor growth inhibition. Combination therapies with adriamycin + cyclophosphamide in PDX mice reduced tumor growth in four Luminal A PDX models. These preclinical results suggest that a two-drug regimen than a three-drug regimen can be useful for breast cancer patients. This study provides insights for future studies combining chemotherapeutics with targeted therapies using PDX models by gamma-irradiation-induced immune suppression.
Collapse
Affiliation(s)
- Solmaz Khalighfard
- Radiation Oncology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Mohammad Alizadeh
- Breast Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran; Cancer Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Amirhoushang Poorkhani
- Clinical Research Development Unit (CRDU), Sayad Shirazi Hospital, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohammadmehdi Motahari
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Arash Tahmasebifar
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ramesh Omranipour
- Breast Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Pedram Keshavarz
- Department of Radiology, Tbilisi State Medical University (TSMU), Tbilisi, Georgia
| | - Peiman Haddad
- Radiation Oncology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
118
|
Patient-derived xenograft (PDX) models of colorectal carcinoma (CRC) as a platform for chemosensitivity and biomarker analysis in personalized medicine. Neoplasia 2020; 23:21-35. [PMID: 33212364 PMCID: PMC7680704 DOI: 10.1016/j.neo.2020.11.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/03/2020] [Accepted: 11/03/2020] [Indexed: 12/16/2022] Open
Abstract
Patient-derived xenograft (PDX) tumor models represent a valuable platform for identifying new biomarkers and novel targets, to evaluate therapy response and resistance mechanisms. This study aimed at establishment, characterization and therapy testing of colorectal carcinoma-derived PDX. We generated 49 PDX and validated identity between patient tumor and corresponding PDX. Sensitivity of PDX toward conventional and targeted drugs revealed that 92% of PDX responded toward irinotecan, 45% toward 5-FU, 65% toward bevacizumab, and 61% toward cetuximab. Expression of epidermal growth factor receptor (EGFR) ligands correlated to the sensitivity toward cetuximab. Proto-oncogene B-RAF, EGFR, Kirsten rat sarcoma virus oncogene homolog gene copy number correlated positively with cetuximab and erlotinib sensitivity. The mutational analyses revealed an individual mutational profile of PDX and mainly identical profiles of PDX from primary tumor vs corresponding metastasis. Mutation in PIK3CA was a determinant of accelerated tumor doubling time. PDX with wildtype Kirsten rat sarcoma virus oncogene homolog, proto-oncogene B-RAF, and phosphatidylinositol-4,5-bisphosphate 3-kinaseM catalytic subunit alfa showed higher sensitivity toward cetuximab and erlotinib. To study the molecular mechanism of cetuximab resistance, cetuximab resistant PDX models were generated, and changes in HER2, HER3, betacellulin, transforming growth factor alfa were observed. Global proteome and phosphoproteome profiling showed a reduction in canonical EGFR-mediated signaling via PTPN11 (SHP2) and AKT1S1 (PRAS40) and an increase in anti-apoptotic signaling as a consequence of acquired cetuximab resistance. This demonstrates that PDX models provide a multitude of possibilities to identify and validate biomarkers, signaling pathways and resistance mechanisms for clinically relevant improvement in cancer therapy.
Collapse
|
119
|
Rashid NU, Luckett DJ, Chen J, Lawson MT, Wang L, Zhang Y, Laber EB, Liu Y, Yeh JJ, Zeng D, Kosorok MR. High-Dimensional Precision Medicine From Patient-Derived Xenografts. J Am Stat Assoc 2020; 116:1140-1154. [PMID: 34548714 PMCID: PMC8451968 DOI: 10.1080/01621459.2020.1828091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 08/28/2020] [Accepted: 09/18/2020] [Indexed: 12/26/2022]
Abstract
The complexity of human cancer often results in significant heterogeneity in response to treatment. Precision medicine offers the potential to improve patient outcomes by leveraging this heterogeneity. Individualized treatment rules (ITRs) formalize precision medicine as maps from the patient covariate space into the space of allowable treatments. The optimal ITR is that which maximizes the mean of a clinical outcome in a population of interest. Patient-derived xenograft (PDX) studies permit the evaluation of multiple treatments within a single tumor, and thus are ideally suited for estimating optimal ITRs. PDX data are characterized by correlated outcomes, a high-dimensional feature space, and a large number of treatments. Here we explore machine learning methods for estimating optimal ITRs from PDX data. We analyze data from a large PDX study to identify biomarkers that are informative for developing personalized treatment recommendations in multiple cancers. We estimate optimal ITRs using regression-based (Q-learning) and direct-search methods (outcome weighted learning). Finally, we implement a superlearner approach to combine multiple estimated ITRs and show that the resulting ITR performs better than any of the input ITRs, mitigating uncertainty regarding user choice. Our results indicate that PDX data are a valuable resource for developing individualized treatment strategies in oncology. Supplementary materials for this article are available online.
Collapse
Affiliation(s)
- Naim U. Rashid
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Daniel J. Luckett
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jingxiang Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Michael T. Lawson
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Longshaokan Wang
- Department of Statistics, North Carolina State University, Raleigh, NC, USA
| | - Yunshu Zhang
- Department of Statistics, North Carolina State University, Raleigh, NC, USA
| | - Eric B. Laber
- Department of Statistics, North Carolina State University, Raleigh, NC, USA
| | - Yufeng Liu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jen Jen Yeh
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Donglin Zeng
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Michael R. Kosorok
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
120
|
Liang C, Bai X, Qi C, Sun Q, Han X, Lan T, Zhang H, Zheng X, Liang R, Jiao J, Zheng Z, Fang J, Lei P, Wang Y, Möckel D, Metselaar JM, Storm G, Hennink WE, Kiessling F, Wei H, Lammers T, Shi Y, Wei B. Π electron-stabilized polymeric micelles potentiate docetaxel therapy in advanced-stage gastrointestinal cancer. Biomaterials 2020; 266:120432. [PMID: 33069116 DOI: 10.1016/j.biomaterials.2020.120432] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 09/10/2020] [Accepted: 10/03/2020] [Indexed: 12/12/2022]
Abstract
Gastrointestinal (GI) cancers are among the most lethal malignancies. The treatment of advanced-stage GI cancer involves standard chemotherapeutic drugs, such as docetaxel, as well as targeted therapeutics and immunomodulatory agents, all of which are only moderately effective. We here show that Π electron-stabilized polymeric micelles based on PEG-b-p(HPMAm-Bz) can be loaded highly efficiently with docetaxel (loading capacity up to 23 wt%) and potentiate chemotherapy responses in multiple advanced-stage GI cancer mouse models. Complete cures and full tumor regression were achieved upon intravenously administering micellar docetaxel in subcutaneous gastric cancer cell line-derived xenografts (CDX), as well as in CDX models with intraperitoneal and lung metastases. Nanoformulated docetaxel also outperformed conventional docetaxel in a patient-derived xenograft (PDX) model, doubling the extent of tumor growth inhibition. Furthermore, micellar docetaxel modulated the tumor immune microenvironment in CDX and PDX tumors, increasing the ratio between M1-and M2-like macrophages, and toxicologically, it was found to be very well-tolerated. These findings demonstrate that Π electron-stabilized polymeric micelles loaded with docetaxel hold significant potential for the treatment of advanced-stage GI cancers.
Collapse
Affiliation(s)
- Chenghua Liang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xiangyang Bai
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany
| | - Cuiling Qi
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Qingxue Sun
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany
| | - Xiaoyan Han
- Central Laboratory, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Tianyun Lan
- Central Laboratory, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Haibo Zhang
- Central Laboratory, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xiaoming Zheng
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Rongpu Liang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Ju Jiao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Zongheng Zheng
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Jiafeng Fang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Purun Lei
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yan Wang
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, the Netherlands
| | - Diana Möckel
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany
| | - Josbert M Metselaar
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, the Netherlands; Department of Biomaterials Science & Technology (BST), University of Twente, 7500 AE, Enschede, the Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, the Netherlands
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany; Fraunhofer MEVIS, Institute for Medical Image Computing, 52074, Aachen, Germany
| | - Hongbo Wei
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, the Netherlands; Department of Biomaterials Science & Technology (BST), University of Twente, 7500 AE, Enschede, the Netherlands.
| | - Yang Shi
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany.
| | - Bo Wei
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
121
|
Rajan V, Melong N, Wong WH, King B, Tong RS, Mahajan N, Gaston D, Lund T, Rittenberg D, Dellaire G, Campbell CJ, Druley T, Berman JN. Humanized zebrafish enhance human hematopoietic stem cell survival and promote acute myeloid leukemia clonal diversity. Haematologica 2020; 105:2391-2399. [PMID: 33054079 PMCID: PMC7556680 DOI: 10.3324/haematol.2019.223040] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 12/05/2019] [Indexed: 11/25/2022] Open
Abstract
Xenograft models are invaluable tools in establishing the current paradigms of hematopoiesis and leukemogenesis. The zebrafish has emerged as a robust alternative xenograft model but, like mice, lack specific cytokines that mimic the microenvironment found in human patients. To address this critical gap, we generated the first humanized zebrafish that express human hematopoietic-specific cytokines (GM-CSF, SCF, and SDF1α). Termed GSS fish, these zebrafish promote survival, self-renewal and multilineage differentiation of human hematopoietic stem and progenitor cells and result in enhanced proliferation and hematopoietic niche-specific homing of primary human leukemia cells. Using error-corrected RNA sequencing, we determined that patient-derived leukemias transplanted into GSS zebrafish exhibit broader clonal representation compared to transplants into control hosts. GSS zebrafish incorporating error-corrected RNA sequencing establish a new standard for zebrafish xenotransplantation that more accurately recapitulates the human context, providing a more representative cost-effective preclinical model system for evaluating personalized response-based treatment in leukemia and therapies to expand human hematopoietic stem and progenitor cells in the transplant setting.
Collapse
Affiliation(s)
- Vinothkumar Rajan
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Nicole Melong
- Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
| | - Wing Hing Wong
- Department of Pediatrics, Division of Hematology-Oncology, Washington University, St. Louis, MO, USA
| | - Benjamin King
- Department of Ocean Sciences, Memorial University, St. John’s, Newfoundland and Labrador, Canada
| | - R. Spencer Tong
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Nitin Mahajan
- Department of Pediatrics, Division of Hematology-Oncology, Washington University, St. Louis, MO, USA
| | - Daniel Gaston
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Troy Lund
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - David Rittenberg
- Department of Obstetrics and Gynecology, IWK Health Science Center, Halifax, Nova Scotia, Canada
| | - Graham Dellaire
- Departments of Pathology and Biochemistry & Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Clinton J.V. Campbell
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada and
| | - Todd Druley
- Department of Pediatrics, Division of Hematology-Oncology, Washington University, St. Louis, MO, USA
| | - Jason N. Berman
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
- CHEO Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
122
|
Huo KG, D'Arcangelo E, Tsao MS. Patient-derived cell line, xenograft and organoid models in lung cancer therapy. Transl Lung Cancer Res 2020; 9:2214-2232. [PMID: 33209645 PMCID: PMC7653147 DOI: 10.21037/tlcr-20-154] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lung cancer accounts for most cancer-related deaths worldwide and has an overall 5-year survival rate of ~15%. Cell lines have played important roles in the study of cancer biology and potential therapeutic targets, as well as pre-clinical testing of novel drugs. However, most experimental therapies that have cleared preclinical testing using established cell lines have failed phase III clinical trials. This suggests that such models may not adequately recapitulate patient tumor biology and clinical outcome predictions. Here, we discuss and compare different pre-clinical lung cancer models, including established cell lines, patient-derived cell lines, xenografts and organoids, summarize the methodology for generating these models, and review their relative advantages and limitations in different oncologic research applications. We further discuss additional gaps in patient-derived pre-clinical models to better recapitulate tumor biology and improve their clinical predictive power.
Collapse
Affiliation(s)
- Ku-Geng Huo
- University Health Network and Princess Margaret Cancer Centre, Toronto, Canada
| | - Elisa D'Arcangelo
- University Health Network and Princess Margaret Cancer Centre, Toronto, Canada
| | - Ming-Sound Tsao
- University Health Network and Princess Margaret Cancer Centre, Toronto, Canada
| |
Collapse
|
123
|
Fortuna-Costa A, Granato RA, Meohas W, Lopes ACDS, Caruso AC, Castro E Silva Pinheiro R, d'Eça PDG, Dias RB, Perini JA, Barbosa APF, Moreira de Sá RA, Guimarães JAM, Murray SS, Duarte MEL. An association between successful engraftment of osteosarcoma patient-derived xenografts and clinicopathological findings. Histol Histopathol 2020; 35:1295-1307. [PMID: 32964941 DOI: 10.14670/hh-18-256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although osteosarcoma is a rare disease, with a global incidence rate estimated at 5.0/million/year, it is the most frequent primary bone sarcoma in children and adolescents. In translational research, the patient-derived xenograft (PDX) model is considered an authentic in vivo model for several types of cancer, as tumorgrafts faithfully retain the biological characteristics of the primary tumors. Our goal was to investigate the association between PDX formation and clinical findings of osteosarcoma patients and the ability of the model to preserve in immunocompromized mice the characteristics of the parental tumor. A fresh sample of the patient tumor obtained from a representative biopsy or from surgical resection was implanted into nude mice. When tumor outgrowths reached ~1,500mm³, fresh PDX fragments were re-transplanted into new hosts. Engraftment in mice was obtained after a latency period of 19-225 days (median 92 days) in 40.54% of the implanted samples. We confirmed the histopathological fidelity between the patient tumor and their respective established PDXs, including the expression of biomarkers. PDX take rate was higher in surgical resection samples, in post-chemotherapy surgical samples and in samples from patients with metastatic disease at presentation. In conclusion, we have shown that the osteosarcoma PDX model reliably recapitulates the morphological aspects of the human disease after serial passage in mice. The observation that more aggressive forms of osteosarcoma, including those with metastatic disease at presentation, have a higher efficiency to generate PDXs provides a promising scenario to address several unanswered issues in clinical oncology.
Collapse
Affiliation(s)
- Anneliese Fortuna-Costa
- Research Division, National Institute of Orthopedics and Traumatology (INTO), Rio de Janeiro, RJ, Brazil
| | | | - Walter Meohas
- Center of Orthopedic Oncology, National Institute of Orthopedics and Traumatology (INTO), Rio de Janeiro, RJ, Brazil
| | - Ana Cristina de Sá Lopes
- Center of Orthopedic Oncology, National Institute of Orthopedics and Traumatology (INTO), Rio de Janeiro, RJ, Brazil
| | - Anabela Cunha Caruso
- Research Division, National Institute of Orthopedics and Traumatology (INTO), Rio de Janeiro, RJ, Brazil
| | | | - Pedro da Gama d'Eça
- Center of Orthopedic Oncology, National Institute of Orthopedics and Traumatology (INTO), Rio de Janeiro, RJ, Brazil
| | - Rhayra Braga Dias
- Research Division, National Institute of Orthopedics and Traumatology (INTO), Rio de Janeiro, RJ, Brazil
| | - Jamila Alessandra Perini
- Research Division, National Institute of Orthopedics and Traumatology (INTO), Rio de Janeiro, RJ, Brazil
| | | | | | | | - Samuel S Murray
- Department of Medicine, University of California, Los Angeles and VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Maria Eugenia Leite Duarte
- Research Division, National Institute of Orthopedics and Traumatology (INTO), Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
124
|
Hu Y, Mi Y, Mukherjee P, Pan Y. A new method of cryopreserving colorectal carcinoma cells for patient derived xenograft model generation. Cryobiology 2020; 96:45-49. [PMID: 32861699 DOI: 10.1016/j.cryobiol.2020.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 12/24/2022]
Abstract
Patient derived xenograft (PDX) models provide an efficient way to study anti-tumor drug efficacy. In this respect, it is essential to study the optimal method needed to cryopreserve the starting cells obtained from tumor samples for PDX model generation. Cryopreservation of cells prior to xenografting is necessary for cross-verification of results obtained by xenografting and also for practical planning of experiments. In the present work, we studied the cryopreservation of colorectal carcinoma (CRC) cells isolated from patient tumor samples for generating their patient derived xenograft models. CRC therapeutics study is essential for early stage intervention and treatment of the disease. CRC cell lines do not ideally depict the molecular characteristics of patient CRC tumor samples. This necessitates the generation of CRC PDX models for drug discovery. We show that CRC cells isolated from patient tumor samples have comparable recovery, viability and growth with both conventional cryopreservation methods as well as Fibulas BioFlash Drive™. However, xenograft tumor formation was much more effective with Fibulas BioFlash Drive™ cryopreserved cells than with cells cryopreserved with conventional methods. Therefore, we put forward an effective way to cryopreserve primary cells obtained from patient tumor samples for PDX model generation in this study.
Collapse
Affiliation(s)
- Yibing Hu
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, 518000, China; Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yulong Mi
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pooja Mukherjee
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94704, United States; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, 94704, United States.
| | - Ying Pan
- CITRIS and the Banatao Institute,330 Sutardja Dai Hall, University of California, Berkeley, Berkeley, CA, 94720-1764, United States.
| |
Collapse
|
125
|
Choi J, Ko EJ, Ju EJ, Park SS, Park J, Shin SH, Jang SJ, Lee JS, Song SY, Jeong SY, Choi EK. Characterization of sphere cells derived from a patient-derived xenograft model of lung adenocarcinoma treated with ionizing radiation. Int J Radiat Biol 2020; 96:1413-1422. [PMID: 32856972 DOI: 10.1080/09553002.2020.1793019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Cancer stem cells (CSCs) are relatively resistant to radiation compared to their non-tumorigenic progeny. Ionizing radiation (IR) can expand the pool of CSCs that leads to more aggressive cancers, but the reason underlying CSC-induced cancer aggressiveness after radiation therapy remains unclear. To understand this, we investigated the phenotypic and molecular characteristics of sphere cells formed from IR-treated patient-derived xenograft (PDX) lung adenocarcinoma tumors. MATERIALS AND METHODS After treatment with various modes of IR, we collected tumors from PDX mice and successfully obtained sphere cells. To compare tumorigenicity, we performed migration, invasion, and mouse transplantation assays with sphere cells from each group. To investigate the molecular features, we used a cDNA microarray and compared gene expression among groups. RESULTS AND CONCLUSIONS Tumorigenicity assays revealed that sphere cells from 2- or 5-Gy IR-treated tumors more aggressive than sphere cells from non-IR treated tumors. Microarray results showed that SERPIB4 and CCL2 were upregulated in sphere cells from IR-treated tumors compared to that in sphere cells from non-IR treated tumors. Interestingly, these genes are related to immune reactions in cancer. Taken together, our results suggest that the aggressiveness of sphere cells obtained after IR treatment is related to resistance, and provide new opportunities for exploring targeted therapies to overcome common radioresistance.
Collapse
Affiliation(s)
- Jinhyang Choi
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Medical Science, University of Ulsan College of Medicine, Seoul, Korea
| | - Eun Jung Ko
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Center for Advancing Cancer Therapeutics, Seoul, Korea
| | - Eun Jin Ju
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Center for Advancing Cancer Therapeutics, Seoul, Korea
| | - Seok Soon Park
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Center for Advancing Cancer Therapeutics, Seoul, Korea
| | - Jin Park
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Center for Advancing Cancer Therapeutics, Seoul, Korea
| | - Seol Hwa Shin
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Center for Advancing Cancer Therapeutics, Seoul, Korea
| | - Se Jin Jang
- Center for Advancing Cancer Therapeutics, Seoul, Korea.,Department of Pathology, Asan Medical Center, Seoul, Korea
| | - Jung Shin Lee
- Department of Internal Medicine, Asan Medical Center, Seoul, Korea
| | - Si Yeol Song
- Center for Advancing Cancer Therapeutics, Seoul, Korea.,Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seong-Yun Jeong
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Center for Advancing Cancer Therapeutics, Seoul, Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eun Kyung Choi
- Center for Advancing Cancer Therapeutics, Seoul, Korea.,Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
126
|
Romaniello D, Marrocco I, Belugali Nataraj N, Ferrer I, Drago-Garcia D, Vaknin I, Oren R, Lindzen M, Ghosh S, Kreitman M, Kittel JC, Gaborit N, Bergado Baez G, Sanchez B, Eilam R, Pikarsky E, Paz-Ares L, Yarden Y. Targeting HER3, a Catalytically Defective Receptor Tyrosine Kinase, Prevents Resistance of Lung Cancer to a Third-Generation EGFR Kinase Inhibitor. Cancers (Basel) 2020; 12:cancers12092394. [PMID: 32847130 PMCID: PMC7563838 DOI: 10.3390/cancers12092394] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
Although two growth factor receptors, EGFR and HER2, are amongst the best targets for cancer treatment, no agents targeting HER3, their kinase-defective family member, have so far been approved. Because emergence of resistance of lung tumors to EGFR kinase inhibitors (EGFRi) associates with compensatory up-regulation of HER3 and several secreted forms, we anticipated that blocking HER3 would prevent resistance. As demonstrated herein, a neutralizing anti-HER3 antibody we generated can clear HER3 from the cell surface, as well as reduce HER3 cleavage by ADAM10, a surface metalloproteinase. When combined with a kinase inhibitor and an anti-EGFR antibody, the antibody completely blocked patient-derived xenograft models that acquired resistance to EGFRi. We found that the underlying mechanism involves posttranslational downregulation of HER3, suppression of MET and AXL upregulation, as well as concomitant inhibition of AKT signaling and upregulation of BIM, which mediates apoptosis. Thus, although HER3 is nearly devoid of kinase activity, it can still serve as an effective drug target in the context of acquired resistance. Because this study simulated in animals the situation of patients who develop resistance to EGFRi and remain with no obvious treatment options, the observations presented herein may warrant clinical testing.
Collapse
Affiliation(s)
- Donatella Romaniello
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel; (D.R.); (I.M.); (N.B.N.); (D.D.-G.); (I.V.); (M.L.); (S.G.); (M.K.); (J.C.K.)
| | - Ilaria Marrocco
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel; (D.R.); (I.M.); (N.B.N.); (D.D.-G.); (I.V.); (M.L.); (S.G.); (M.K.); (J.C.K.)
| | - Nishanth Belugali Nataraj
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel; (D.R.); (I.M.); (N.B.N.); (D.D.-G.); (I.V.); (M.L.); (S.G.); (M.K.); (J.C.K.)
| | - Irene Ferrer
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain; (I.F.); (L.P.-A.)
- Lung Cancer Clinical Research Unit, Instituto de Investigación Hospital 12 de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain
| | - Diana Drago-Garcia
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel; (D.R.); (I.M.); (N.B.N.); (D.D.-G.); (I.V.); (M.L.); (S.G.); (M.K.); (J.C.K.)
| | - Itay Vaknin
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel; (D.R.); (I.M.); (N.B.N.); (D.D.-G.); (I.V.); (M.L.); (S.G.); (M.K.); (J.C.K.)
| | - Roni Oren
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 76100, Israel; (R.O.); (R.E.)
| | - Moshit Lindzen
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel; (D.R.); (I.M.); (N.B.N.); (D.D.-G.); (I.V.); (M.L.); (S.G.); (M.K.); (J.C.K.)
| | - Soma Ghosh
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel; (D.R.); (I.M.); (N.B.N.); (D.D.-G.); (I.V.); (M.L.); (S.G.); (M.K.); (J.C.K.)
| | - Matthew Kreitman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel; (D.R.); (I.M.); (N.B.N.); (D.D.-G.); (I.V.); (M.L.); (S.G.); (M.K.); (J.C.K.)
| | - Jeanette Clarissa Kittel
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel; (D.R.); (I.M.); (N.B.N.); (D.D.-G.); (I.V.); (M.L.); (S.G.); (M.K.); (J.C.K.)
| | - Nadege Gaborit
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, 34298 Montpellier, France;
- Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Gretchen Bergado Baez
- Tumor Biology Direction, Center of Molecular Immunology, Havana 11600, Cuba; (G.B.B.); (B.S.)
| | - Belinda Sanchez
- Tumor Biology Direction, Center of Molecular Immunology, Havana 11600, Cuba; (G.B.B.); (B.S.)
| | - Raya Eilam
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 76100, Israel; (R.O.); (R.E.)
| | - Eli Pikarsky
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel;
| | - Luis Paz-Ares
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain; (I.F.); (L.P.-A.)
- Lung Cancer Clinical Research Unit, Instituto de Investigación Hospital 12 de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain
- Medical Oncology Department, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel; (D.R.); (I.M.); (N.B.N.); (D.D.-G.); (I.V.); (M.L.); (S.G.); (M.K.); (J.C.K.)
- Correspondence: ; Tel.: +972-8-934-3974
| |
Collapse
|
127
|
Human-Derived Model Systems in Gynecological Cancer Research. Trends Cancer 2020; 6:1031-1043. [PMID: 32855097 DOI: 10.1016/j.trecan.2020.07.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/24/2022]
Abstract
The human female reproductive tract (FRT) is a complex system that combines series of organs, including ovaries, fallopian tubes, uterus, cervix, vagina, and vulva; each of which possesses unique cellular characteristics and functions. This versatility, in turn, allows for the development of a wide range of epithelial gynecological cancers with distinct features. Thus, reliable model systems are required to better understand the diverse mechanisms involved in the regional pathogenesis of the reproductive tract and improve treatment strategies. Here, we review the current human-derived model systems available to study the multitude of gynecological cancers, including ovarian, endometrial, cervical, vaginal, and vulvar cancer, and the recent advances in the push towards personalized therapy.
Collapse
|
128
|
Palmer AC, Plana D, Gao H, Korn JM, Yang G, Green J, Zhang X, Velazquez R, McLaughlin ME, Ruddy DA, Kowal C, Muszynski J, Bullock C, Rivera S, Rakiec DP, Elliott G, Fordjour P, Meyer R, Loo A, Kurth E, Engelman JA, Bitter H, Sellers WR, Williams JA, Sorger PK. A Proof of Concept for Biomarker-Guided Targeted Therapy against Ovarian Cancer Based on Patient-Derived Tumor Xenografts. Cancer Res 2020; 80:4278-4287. [PMID: 32747364 DOI: 10.1158/0008-5472.can-19-3850] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/29/2020] [Accepted: 07/29/2020] [Indexed: 12/26/2022]
Abstract
Advanced ovarian cancers are a leading cause of cancer-related death in women and are currently treated with surgery and chemotherapy. This standard of care is often temporarily successful but exhibits a high rate of relapse, after which, treatment options are few. Here we investigate whether biomarker-guided use of multiple targeted therapies, including small molecules and antibody-drug conjugates, is a viable alternative. A panel of patient-derived ovarian cancer xenografts (PDX), similar in genetics and chemotherapy responsiveness to human tumors, was exposed to 21 monotherapies and combination therapies. Three monotherapies and one combination were found to be active in different subsets of PDX. Analysis of gene expression data identified biomarkers associated with responsiveness to each of the three targeted therapies, none of which directly inhibits an oncogenic driver. While no single treatment had as high a response rate as chemotherapy, nearly 90% of PDXs were eligible for and responded to at least one biomarker-guided treatment, including tumors resistant to standard chemotherapy. The distribution of biomarker positivity in The Cancer Genome Atlas data suggests the potential for a similar precision approach in human patients. SIGNIFICANCE: This study exploits a panel of patient-derived xenografts to demonstrate that most ovarian tumors can be matched to effective biomarker-guided treatments.
Collapse
Affiliation(s)
- Adam C Palmer
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, Massachusetts.,Department of Pharmacology, Computational Medicine Program, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Deborah Plana
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, Massachusetts.,Department of Systems Biology, Harvard Medical School, Boston, Massachusetts.,Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School and MIT, Cambridge, Massachusetts
| | - Hui Gao
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Joshua M Korn
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Guizhi Yang
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - John Green
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Xiamei Zhang
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Roberto Velazquez
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Margaret E McLaughlin
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - David A Ruddy
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Colleen Kowal
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Julie Muszynski
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Caroline Bullock
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Stacy Rivera
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Daniel P Rakiec
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - GiNell Elliott
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Paul Fordjour
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Ronald Meyer
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Alice Loo
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Esther Kurth
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Jeffrey A Engelman
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Hans Bitter
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - William R Sellers
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Juliet A Williams
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts.
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, Massachusetts. .,Department of Systems Biology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
129
|
Wu Z, Gong Z, Ao Z, Xu J, Cai H, Muhsen M, Heaps S, Bondesson M, Guo S, Guo F. Rapid Microfluidic Formation of Uniform Patient-Derived Breast Tumor Spheroids. ACS APPLIED BIO MATERIALS 2020; 3:6273-6283. [DOI: 10.1021/acsabm.0c00768] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Zhuhao Wu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Zhiyi Gong
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Zheng Ao
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Junhua Xu
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Maram Muhsen
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Samuel Heaps
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Maria Bondesson
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Shishang Guo
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
130
|
Knier NN, Hamilton AM, Foster PJ. Comparing the fate of brain metastatic breast cancer cells in different immune compromised mice with cellular magnetic resonance imaging. Clin Exp Metastasis 2020; 37:465-475. [PMID: 32533389 DOI: 10.1007/s10585-020-10044-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/05/2020] [Indexed: 01/28/2023]
Abstract
Metastasis is the leading cause of mortality in breast cancer patients, with brain metastases becoming increasingly prevalent. Studying this disease is challenging due to the limited experimental models and methods available. Here, we used iron-based cellular MRI to track the fate of a mammary carcinoma cell line (MDA-MB-231-BR) in vivo to characterize the growth of brain metastases in the nude and severely immune-compromised NOD/SCID/ILIIrg-/- (NSG) mouse. Nude and NSG mice received injections of iron-labeled MDA-MB-231-BR cells. Images were acquired with a 3T MR system and assessed for signal voids and metastases. The percentage of signal voids and the number and volume of metastases were quantified. Ex vivo imaging of the liver, histology, and immunofluorescence labeling was performed. Brain metastases grew more rapidly in NSG mice. At day 21 post cell injection, the average number of brain tumors in NSG mice was approximately four times greater than in nude mice. The persistence of iron-labeled cells, visualized as signal voids by MRI, was also examined. The percentage of voids decreased significantly over time for both nude and NSG mice. Body images revealed that the NSG mice also had metastases in the liver, lungs, and lymph nodes while tumors were only detected in the brains of nude mice. This work demonstrates the advantages of using the highly immune-compromised NSG mouse to study breast cancer metastasis, treatments aimed at inhibiting metastasis and outgrowth of breast cancer metastases in multiple organs, and the role that imaging can play toward credentialing these models that cannot be done with other in vitro or histopathologic methods alone.
Collapse
Affiliation(s)
- Natasha N Knier
- Imaging Research Laboratories, Robarts Research Institute, 1151 Richmond St. N., London, ON, N6A 5B7, Canada. .,Department of Medical Biophysics, Western University, 1151 Richmond St, London, ON, N6A 3K7, Canada.
| | - Amanda M Hamilton
- Imaging Research Laboratories, Robarts Research Institute, 1151 Richmond St. N., London, ON, N6A 5B7, Canada
| | - Paula J Foster
- Imaging Research Laboratories, Robarts Research Institute, 1151 Richmond St. N., London, ON, N6A 5B7, Canada.,Department of Medical Biophysics, Western University, 1151 Richmond St, London, ON, N6A 3K7, Canada
| |
Collapse
|
131
|
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: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/13/2022]
|
132
|
Wu X, Wang S, Li M, Li J, Shen J, Zhao Y, Pang J, Wen Q, Chen M, Wei B, Kaboli PJ, Du F, Zhao Q, Cho CH, Wang Y, Xiao Z, Wu X. Conditional reprogramming: next generation cell culture. Acta Pharm Sin B 2020; 10:1360-1381. [PMID: 32963937 PMCID: PMC7488362 DOI: 10.1016/j.apsb.2020.01.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
Long-term primary culture of mammalian cells has been always difficult due to unavoidable senescence. Conventional methods for generating immortalized cell lines usually require manipulation of genome which leads to change of important biological and genetic characteristics. Recently, conditional reprogramming (CR) emerges as a novel next generation tool for long-term culture of primary epithelium cells derived from almost all origins without alteration of genetic background of primary cells. CR co-cultures primary cells with inactivated mouse 3T3-J2 fibroblasts in the presence of RHO-related protein kinase (ROCK) inhibitor Y-27632, enabling primary cells to acquire stem-like characteristics while retain their ability to fully differentiate. With only a few years' development, CR shows broad prospects in applications in varied areas including disease modeling, regenerative medicine, drug evaluation, drug discovery as well as precision medicine. This review is thus to comprehensively summarize and assess current progress in understanding mechanism of CR and its wide applications, highlighting the value of CR in both basic and translational researches and discussing the challenges faced with CR.
Collapse
Key Words
- 3T3-J2 fibroblast
- AACR, American Association for Cancer Research
- ACC, adenoid cystic carcinoma
- AR, androgen receptor
- CFTR, cystic fibrosis transmembrane conductance regulators
- CR, conditional reprogramming
- CYPs, cytochrome P450 enzymes
- Conditional reprogramming
- DCIS, ductal carcinoma in situ
- ECM, extracellular matrix
- ESC, embryonic stem cell
- HCMI, human cancer model initiatives
- HGF, hepatocyte growth factor
- HNE, human nasal epithelial
- HPV, human papillomaviruses
- ICD, intracellular domain
- LECs, limbal epithelial cells
- NCI, National Cancer Institute
- NGFR, nerve growth factor receptor
- NSCLC, non-small cell lung cancer
- NSG, NOD/SCID/gamma
- PDAC, pancreatic ductal adenocarcinoma
- PDX, patient derived xenograft
- PP2A, protein phosphatase 2A
- RB, retinoblastoma-associated protein
- ROCK
- ROCK, Rho kinase
- SV40, simian virus 40 large tumor antigen
- Senescence
- UVB, ultraviolet radiation b
- Y-27632
- dECM, decellularized extracellular matrix
- hASC, human adipose stem cells
- hTERT, human telomerase reverse transcriptase
- iPSCs, induction of pluripotent stem cells
- ΔNP63α, N-terminal truncated form of P63α
Collapse
Affiliation(s)
- Xiaoxiao Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Jing Li
- Department of Oncology and Hematology, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou 646000, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Jun Pang
- Center of Radiation Oncology, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou 646000, China
| | - Qinglian Wen
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou 646000, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Bin Wei
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Parham Jabbarzadeh Kaboli
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Qijie Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| |
Collapse
|
133
|
Ehmsen S, Pedersen MH, Wang G, Terp MG, Arslanagic A, Hood BL, Conrads TP, Leth-Larsen R, Ditzel HJ. Increased Cholesterol Biosynthesis Is a Key Characteristic of Breast Cancer Stem Cells Influencing Patient Outcome. Cell Rep 2020; 27:3927-3938.e6. [PMID: 31242424 DOI: 10.1016/j.celrep.2019.05.104] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 04/01/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022] Open
Abstract
Tumor eradication may be greatly improved by targeting cancer stem cells (CSCs), as they exhibit resistance to conventional therapy. To gain insight into the unique biology of CSCs, we developed patient-derived xenograft tumors (PDXs) from ER- breast cancers from which we isolated mammospheres that are enriched for CSCs. Comparative global proteomic analysis was performed on patient tumor tissues and corresponding PDXs and mammospheres. Mammospheres exhibited increased expression of proteins associated with de novo cholesterol synthesis. The clinical relevance of increased cholesterol biosynthesis was verified in a large breast cancer cohort showing correlation with shorter relapse-free survival. RNAi and chemical inhibition of the cholesterol biosynthesis pathway reduced mammosphere formation, which could be rescued by a downstream metabolite. Our findings identify the cholesterol biosynthesis pathway as central for CSC propagation and a potential therapeutic target, as well as providing a mechanistic explanation for the therapeutic benefit of statins in breast cancer.
Collapse
Affiliation(s)
- Sidse Ehmsen
- Department of Molecular Medicine, Cancer, and Inflammation Research Unit, University of Southern Denmark, 5000 Odense, Denmark; Department of Oncology, Odense University Hospital, 5000 Odense, Denmark; Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, 5000 Odense, Denmark
| | - Martin H Pedersen
- Department of Molecular Medicine, Cancer, and Inflammation Research Unit, University of Southern Denmark, 5000 Odense, Denmark
| | - Guisong Wang
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Annandale, VA 22003, USA
| | - Mikkel G Terp
- Department of Molecular Medicine, Cancer, and Inflammation Research Unit, University of Southern Denmark, 5000 Odense, Denmark
| | - Amina Arslanagic
- Department of Molecular Medicine, Cancer, and Inflammation Research Unit, University of Southern Denmark, 5000 Odense, Denmark
| | - Brian L Hood
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Annandale, VA 22003, USA
| | - Thomas P Conrads
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Annandale, VA 22003, USA; Inova Schar Cancer Institute, Inova Center for Personalized Health, Fairfax, VA 22031, USA
| | - Rikke Leth-Larsen
- Department of Molecular Medicine, Cancer, and Inflammation Research Unit, University of Southern Denmark, 5000 Odense, Denmark; Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark.
| | - Henrik J Ditzel
- Department of Molecular Medicine, Cancer, and Inflammation Research Unit, University of Southern Denmark, 5000 Odense, Denmark; Department of Oncology, Odense University Hospital, 5000 Odense, Denmark; Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark; Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, 5000 Odense, Denmark.
| |
Collapse
|
134
|
Beyond the Genomic Mutation: Rethinking the Molecular Biomarkers of K-RAS Dependency in Pancreatic Cancers. Int J Mol Sci 2020; 21:ijms21145023. [PMID: 32708716 PMCID: PMC7404119 DOI: 10.3390/ijms21145023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Oncogenic v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (K-RAS) plays a key role in the development and maintenance of pancreatic ductal adenocarcinoma (PDAC). The targeting of K-RAS would be beneficial to treat tumors whose growth depends on active K-RAS. The analysis of K-RAS genomic mutations is a clinical routine; however, an emerging question is whether the mutational status is able to identify tumors effectively dependent on K-RAS for tailoring targeted therapies. With the emergence of novel K-RAS inhibitors in clinical settings, this question is relevant. Several studies support the notion that the K-RAS mutation is not a sufficient biomarker deciphering the effective dependency of the tumor. Transcriptomic and metabolomic profiles of tumors, while revealing K-RAS signaling complexity and K-RAS-driven molecular pathways crucial for PDAC growth, are opening the opportunity to specifically identify K-RAS-dependent- or K-RAS-independent tumor subtypes by using novel molecular biomarkers. This would help tumor selection aimed at tailoring therapies against K-RAS. In this review, we will present studies about how the K-RAS mutation can also be interpreted in a state of K-RAS dependency, for which it is possible to identify specific K-RAS-driven molecular biomarkers in certain PDAC subtypes, beyond the genomic K-RAS mutational status.
Collapse
|
135
|
Shi J, Zhang Y, Wang J, Li J, Li Z. Anlotinib Combined With Chemoradiotherapy Exhibits Significant Therapeutic Efficacy in Esophageal Squamous Cell Carcinoma. Front Oncol 2020; 10:995. [PMID: 32754439 PMCID: PMC7365939 DOI: 10.3389/fonc.2020.00995] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
Objective: The aim of this study was to evaluate the safety and efficacy of anlotinib combined with chemoradiotherapy for treating esophageal squamous cell carcinoma (ESCC) using patient-derived xenografts (PDXs). Methods: PDX-bearing mice were randomly divided into five groups, as follows: control group receiving normal saline, the group receiving radiotherapy, the group receiving cisplatin combined with radiotherapy, the group receiving anlotinib combined with radiotherapy, and the group receiving anlotinib, and cisplatin combined with radiotherapy. Tumor volumes and body weights were measured three times weekly for 2 weeks. The PDXs were initially assessed by comparing the histology of the original patient tumor tissues with that of the corresponding serially passaged xenografts by hematoxylin and eosin (H&E) and P63 staining. Then, expression of Bax, c-PARP, PCNA, and CD31 was detected using immunohistochemistry, and apoptosis was detected by a TUNEL assay. Cytokines released into plasma were analyzed using protein chip technology. Finally, two case studies of ESCC patients were presented to further verify the results observed in the PDX models. Results: The pathological characteristics of the serially passaged patient tumor-derived xenografts established in our study were in line with those of the original ESCC patient samples. The group receiving anlotinib and cisplatin plus radiotherapy exhibited the strongest antitumor response among the groups. Moreover, the ideal anticancer effects of anlotinib combined with chemoradiotherapy observed in clinical patients were consistent with the results observed in the PDX models, and no serious side effects were observed during treatment. Conclusions: Combination therapy with anlotinib and chemoradiotherapy may be an effective regimen for the treatment of advanced ESCC.
Collapse
Affiliation(s)
- Jingzhen Shi
- School of Medicine, Shandong University, Jinan, China.,Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yingjie Zhang
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jinzhi Wang
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jianbin Li
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhenxiang Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
136
|
Ripsman D, Fergusson DA, Montroy J, Auer RC, Huang JW, Dobriyal A, Wesch N, Carrier M, Lalu MM. A systematic review on the efficacy and safety of low molecular weight heparin as an anticancer therapeutic in preclinical animal models. Thromb Res 2020; 195:103-113. [PMID: 32683148 DOI: 10.1016/j.thromres.2020.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 01/30/2023]
Abstract
OBJECTIVE The therapeutic effects of low molecular weight heparins (LMWH) may extend past thrombosis prevention, with preclinical evidence demonstrating anti-metastatic properties. Clinical evidence on the topic, however, remains controversial. A systematic review of preclinical evidence may help elucidate reasons for this contradictory evidence. The objective of our systematic review is to assess the anti-metastatic properties of LMWHs in solid tumour animal models. METHODS MEDLINE, Embase, Web of Science and PubMed were searched from inception to May 12th, 2020. All articles were screened independently and in duplicate. Studies that compared LMWH to a placebo or no treatment arm in solid tumour animal models were included. The primary outcome was the burden of metastasis. Secondary outcomes included primary tumour growth and mortality. The risk of bias was assessed in duplicate using a modified Cochrane Risk of Bias tool. RESULTS Forty-two studies were included in the review. Administration of a LMWH was associated with a significant decrease in the burden of metastasis (SMD -2.18; 95% CI -2.66 to -1.70). Additionally, the administration of a LMWH was also associated with a significant reduction in primary tumour growth (SMD -1.95; 95% CI -2.56 to -1.34) and risk of death (RR 0.39; 95% CI 0.16-0.97). All included studies were deemed to be at an unclear risk of bias for at least one methodological criterion. CONCLUSIONS Our results demonstrate that LMWH can effectively reduce metastatic burden and reduce tumour growth in preclinical animal models of solid tumour malignancies. Reasons for the contradiction with clinical evidence require further exploration.
Collapse
Affiliation(s)
- David Ripsman
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada; Faculty of Medicine, University of Ottawa, 451 Smyth Rd #2044, Ottawa, ON K1H 8M5, Canada.
| | - Dean A Fergusson
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada; Faculty of Medicine, University of Ottawa, 451 Smyth Rd #2044, Ottawa, ON K1H 8M5, Canada.
| | - Joshua Montroy
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada.
| | - Rebecca C Auer
- Department of Surgery, The Ottawa Hospital, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada; Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada.
| | - Johnny W Huang
- Faculty of Medicine, University of Ottawa, 451 Smyth Rd #2044, Ottawa, ON K1H 8M5, Canada.
| | - Aditi Dobriyal
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada; Faculty of Medicine, University of Ottawa, 451 Smyth Rd #2044, Ottawa, ON K1H 8M5, Canada.
| | - Neil Wesch
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada
| | - Marc Carrier
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada; Faculty of Medicine, University of Ottawa, 451 Smyth Rd #2044, Ottawa, ON K1H 8M5, Canada.
| | - Manoj M Lalu
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada; Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Room B307, 1053 Carling Avenue, Mail Stop 249, Ottawa, ON K1Y 4E9, Canada; Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada.
| |
Collapse
|
137
|
Ex vivo culture of head and neck cancer explants in cell sheet for testing chemotherapeutic sensitivity. J Cancer Res Clin Oncol 2020; 146:2497-2507. [PMID: 32620987 DOI: 10.1007/s00432-020-03306-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/27/2020] [Indexed: 12/26/2022]
Abstract
PURPOSE Tumor explant culture systems can mimic the in vivo tumor microenvironment, proposing as a substitute for preclinical studies for prediction of individual treatment response. Therefore, our study evaluated the potential usefulness of ex vivo tumor explants culture assembled into the cell sheets by anticancer drug screening in patients with head and neck squamous cell carcinoma (HNSCC). METHODS Our model included tumor explants incorporated into cell sheet composing of epithelium and subepithelial stroma using tumor and mucosal samples obtained from the HNSCC patients who underwent surgery. Cell growth, viability, and hypoxia were measured by cell counting kit-8, live/dead assay, propidium iodide, and LOX-1 staining, and were compared among the different treatment groups with vehicle, cisplatin or docetaxel. RESULTS Tumor explants stably survived in the cell sheet over 10 days after explantation, whereas most of the explants in non-matrix culture became nonviable within 5-8 days with the significant daily decrease of viability. The live tissue areas of tumor explants in the cell sheet maintained over 30 days without significant changes although hypoxic cell areas gradually increased up to 5 days. Tissue viability and live cancer tissue areas significantly decreased after the treatment of cisplatin or docetaxel in the dose and time-dependent manners. CONCLUSION Our cell sheet-based tumor explants model might be applied to the reliable ex vivo screening for anticancer chemotherapeutics for HNSCC.
Collapse
|
138
|
Greenhalgh CJ, Karekla E, Miles GJ, Powley IR, Costa C, de Jesus J, Bailey MJ, Pritchard C, MacFarlane M, Pringle JH, Managh AJ. Exploration of Matrix Effects in Laser Ablation Inductively Coupled Plasma Mass Spectrometry Imaging of Cisplatin-Treated Tumors. Anal Chem 2020; 92:9847-9855. [DOI: 10.1021/acs.analchem.0c01347] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Calum J. Greenhalgh
- Department of Chemistry, Loughborough University, Loughborough, Leicestershire LE11 3TU, U.K
| | - Ellie Karekla
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester LE2 7LX, U.K
| | - Gareth J. Miles
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester LE2 7LX, U.K
| | - Ian R. Powley
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester LE2 7LX, U.K
| | - Catia Costa
- Ion Beam Centre, University of Surrey, Guildford GU2 7XH, U.K
| | - Janella de Jesus
- Department of Chemistry, University of Surrey, Guildford GU2 7XH, U.K
| | - Melanie J. Bailey
- Department of Chemistry, University of Surrey, Guildford GU2 7XH, U.K
| | - Catrin Pritchard
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester LE2 7LX, U.K
| | | | - J. Howard Pringle
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester LE2 7LX, U.K
| | - Amy J. Managh
- Department of Chemistry, Loughborough University, Loughborough, Leicestershire LE11 3TU, U.K
| |
Collapse
|
139
|
Zu X, Ma X, Xie X, Lu B, Laster K, Liu K, Dong Z, Kim DJ. 2,6-DMBQ is a novel mTOR inhibitor that reduces gastric cancer growth in vitro and in vivo. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:107. [PMID: 32517736 PMCID: PMC7285595 DOI: 10.1186/s13046-020-01608-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022]
Abstract
Background Fermented wheat germ extract has been reported to exert various pharmacological activities, including anti-oxidant, anti-cell growth and cell apoptosis in various cancer cells. Although 2,6-dimethoxy-1,4-benzoquinone (2,6-DMBQ) is a benzoquinone compound and found in fermented wheat germ extract, its anticancer effects and molecular mechanism(s) against gastric cancer have not been investigated. Methods Anticancer effects of 2,6-DMBQ were determined by MTT, soft agar, cell cycle and Annexin V analysis. Potential candidate proteins were screened via in vitro kinase assay and Western blotting. mTOR knockdown cell lines were established by lentiviral infection with shmTOR. The effect of 2,6-DMBQ on tumor growth was assessed using gastric cancer patient-derived xenograft models. Results 2,6-DMBQ significantly reduced cell growth and induced G1 phase cell cycle arrest and apoptosis in gastric cancer cells. 2,6-DMBQ reduced the activity of mTOR in vitro. The inhibition of cell growth by 2,6-DMBQ is dependent upon the expression of the mTOR protein. Remarkably, 2,6-DMBQ strongly reduced patient-derived xenograft gastric tumor growth in an in vivo mouse model. Conclusions 2,6-DMBQ is an mTOR inhibitor that can be useful for treating gastric cancer. It has therapeutic implications for gastric cancer patients.
Collapse
Affiliation(s)
- Xueyin Zu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450008, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China
| | - Xiaoli Ma
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450008, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China
| | - Xiaomeng Xie
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450008, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China
| | - Bingbing Lu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450008, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China
| | - Kyle Laster
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China
| | - Kangdong Liu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450008, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China.,The Affiliated Cancer Hospital, Zhengzhou University, Zhengzhou, 450008, Henan, China.,The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, 450008, Henan, China
| | - Zigang Dong
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450008, Henan, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China. .,The Affiliated Cancer Hospital, Zhengzhou University, Zhengzhou, 450008, Henan, China. .,The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, 450008, Henan, China. .,International joint research center of cancer chemoprevention, Zhengzhou, China.
| | - Dong Joon Kim
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450008, Henan, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China. .,The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, 450008, Henan, China.
| |
Collapse
|
140
|
Chagoya G, Kwatra SG, Nanni CW, Roberts CM, Phillips SM, Nullmeyergh S, Gilmore SP, Spasojevic I, Corcoran DL, Young CC, Ballman KV, Ramakrishna R, Cross DA, Markert JM, Lim M, Gilbert MR, Lesser GJ, Kwatra MM. Efficacy of osimertinib against EGFRvIII+ glioblastoma. Oncotarget 2020; 11:2074-2082. [PMID: 32547705 PMCID: PMC7275784 DOI: 10.18632/oncotarget.27599] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/27/2020] [Indexed: 11/25/2022] Open
Abstract
Epidermal Growth Factor Receptor variant III (EGFRvIII) is an active mutant form of EGFR that drives tumor growth in a subset of glioblastoma (GBM). It occurs in over 20% of GBMs, making it a promising receptor for small molecule targeted therapy. We hypothesize that poor penetration of the blood-brain barrier by previously tested EGFR-tyrosine kinase inhibitors (EGFR-TKIs) such as afateninb, erlotinib, gefitinib, and lapatinib played a role in their limited efficacy. The present study examined the effects of osimertinib (previously known as AZD9291) on EGFRvIII+ GBM models, both in vitro and in vivo. Therefore, a panel of six GBM stem cells (GSCs) expressing EGFRvIII+ was evaluated. The EGFRvIII+ GSC differed in the expression of EGFRvIII and other key genes. The GSC line D317, which expresses high levels of EGFRvIII and has robust tyrosine kinase activity, was selected for assessing osimertinib’s efficacy. Herein, we report that osimertinib inhibits the constitutive activity of EGFRvIII tyrosine kinase with high potency (<100 nM) while also inhibiting its downstream signaling. Further, osimertinib inhibited D317’s growth in vitro and in both heterotopic and orthotopic xenograft models. Additional preclinical studies are warranted to identify EGFRvIII+ GBM’s molecular signature most responsive to osimertinib.
Collapse
Affiliation(s)
- Gustavo Chagoya
- Department of Neurosurgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shawn G Kwatra
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cory W Nanni
- Departments of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Callie M Roberts
- Departments of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Samantha M Phillips
- Tri-Institutional MD-PhD Program, Weill Cornell Medical College, The Rockefeller University, Memorial Sloan Kettering Cancer Institute, New York, NY, USA
| | - Sarah Nullmeyergh
- Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Samuel P Gilmore
- Departments of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Ivan Spasojevic
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - David L Corcoran
- Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Christopher C Young
- Departments of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Karla V Ballman
- Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, NY, USA
| | | | - Darren A Cross
- IMED Oncology, Global Medical Affairs, AstraZeneca, Cambridge, UK
| | - James M Markert
- Department of Neurosurgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael Lim
- Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Glenn J Lesser
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Madan M Kwatra
- Departments of Anesthesiology, Duke University Medical Center, Durham, NC, USA.,Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA.,Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
141
|
Fang B, Kannan A, Zhao S, Nguyen QH, Ejadi S, Yamamoto M, Camilo Barreto J, Zhao H, Gao L. Inhibition of PI3K by copanlisib exerts potent antitumor effects on Merkel cell carcinoma cell lines and mouse xenografts. Sci Rep 2020; 10:8867. [PMID: 32483262 PMCID: PMC7264292 DOI: 10.1038/s41598-020-65637-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
Merkel cell carcinoma (MCC) is a highly aggressive neuroendocrine skin cancer with steadily increasing incidence and poor prognosis. Despite recent success with immunotherapy, 50% of patients still succumb to their diseases. To date, there is no Food and Drug Administration-approved targeted therapy for advanced MCC. Aberrant activation of phosphatidylinositide-3-kinase (PI3K)/AKT/mTOR pathway is frequently detected in MCC, making it an attractive therapeutic target. We previously found PI3K pathway activation in human MCC cell lines and tumors and demonstrated complete clinical response in a Stage IV MCC patient treated with PI3K inhibitor idelalisib. Here, we found that both PI3K-α and -δ isoforms are abundantly expressed in our MCC cell lines and clinical samples; we therefore examined antitumor efficacy across a panel of five PI3K inhibitors with distinctive isoform-specificities, including idelalisib (PI3K-δ), copanlisib (PI3K-α/δ), duvelisib (PI3K-γ/δ), alpelisib (PI3K-α), and AZD8186 (PI3K-β/δ). Of these, copanlisib exerts the most potent antitumor effects, markedly inhibiting cell proliferation, survival, and tumor growth by suppressing PI3K/mTOR/Akt activities in mouse models generated from MCC cell xenografts and patient-derived tumor xenografts. These results provide compelling preclinical evidence for application of copanlisib in advanced MCC with aberrant PI3K activation for which immunotherapy is insufficient, or patients who are unsuitable for immunotherapy.
Collapse
Affiliation(s)
- Bin Fang
- Department of Dermatology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Aarthi Kannan
- Department of Dermatology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Southern California Institute of Research and Education, Long Beach, CA, USA
- Department of Dermatology, University of California - Irvine, Irvine, CA, USA
| | | | - Quy H Nguyen
- Department of Biological Chemistry, University of California - Irvine, Irvine, CA, USA
| | - Samuel Ejadi
- Division of Hematology/Oncology, School of Medicine, University of California - Irvine, Irvine, CA, USA
| | - Maki Yamamoto
- Department of Surgery, School of Medicine, University of California - Irvine, Irvine, CA, USA
| | - J Camilo Barreto
- Department of Surgery, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Haibo Zhao
- Southern California Institute of Research and Education, Long Beach, CA, USA
| | - Ling Gao
- Department of Dermatology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
- Southern California Institute of Research and Education, Long Beach, CA, USA.
- Department of Dermatology, University of California - Irvine, Irvine, CA, USA.
- Veterans Affairs Long Beach Healthcare System, Long Beach, CA, USA.
| |
Collapse
|
142
|
Li W, Zhang X, Xi X, Li Y, Quan H, Liu S, Wu L, Wu P, Lan W, Shao Y, Li H, Chen K, Hu Z. PLK2 modulation of enriched TAp73 affects osteogenic differentiation and prognosis in human osteosarcoma. Cancer Med 2020; 9:4371-4385. [PMID: 32349184 PMCID: PMC7300400 DOI: 10.1002/cam4.3066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/26/2020] [Accepted: 03/29/2020] [Indexed: 02/05/2023] Open
Abstract
There are three subtypes of undifferentiated human conventional osteosarcoma (HCOS): osteoblastic osteosarcoma (OOS), chondroblastic osteosarcoma (COS), and fibroblastic osteosarcoma (FOS). HCOS also exhibits heterogeneous pathological maldifferentiation in individual patients. Currently, the mechanism regulating HCOS differentiation remains unclear, and therapies are ineffective. Osteopontin (OPN) and osteocalcin (OCN) are markers of osteoblast maturation, and their expression is inhibited in HCOS. A previous study found that PLK2 inhibited TAp73 phosphorylation and consequent anti-OS function of TAp73 in OS cells with enriched TAp73. TAp73 was also reported to regulate bone cell calcification. Here, OOS was found to have higher TAp73 levels and PLK2 expression than those in COS, which is correlated with HCOS maldifferentiation according to Spearman analysis and affects patient prognosis according to Kaplan-Meier survival analysis. In the conventional OS cell-line Saos2 and in patient-derived xenograft OS (PDX-OS) cells, increased PLK2 expression owing to abundant TAp73 levels affected OPN and OCN content as measured by RT-PCR and Western blotting, and alizarin red staining showed that PLK2 affected calcium deposition in OS cells. In addition, PLK2 inhibition in PDX-OS cells prohibited clone formation, as indicated by a clonogenic assay, and sensitized OS cells to cisplatin (CDDP) (which consequently limited proliferation), as shown by the CCK-8 assay. In an established PDX animal model with abundant TAp73 levels, PLK2 inhibition or CDDP treatment prevented tumor growth and prolonged median survival. The combined therapeutic effect of PLK2 inhibition with CDDP treatment was better than that of either monotherapy. These results indicate that increased PLK2 levels due to enriched TAp73 affect osteogenic differentiation and maturation and OS prognosis. In conclusion, PLK2 is a potential target for differentiation therapy of OS with enriched TAp73.
Collapse
Affiliation(s)
- Wenhu Li
- Department of OrthopedicsShaoguan First People's Hospital Affiliated to Southern Medical UniversityShaoguanChina
| | - Xianliao Zhang
- Orthopedics CenterZhujiang Hospital of Southern Medical UniversityGuangzhouChina
| | - Xinhua Xi
- Department of OrthopaedicsThe Affiliated Yuebei People's Hospital of Shantou University Medical CollegeShaoguanChina
| | - Yufa Li
- The Second School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
- Department of PathologyGuangdong provincial people's Hospital & Guangdong, Academy of Medical SciencesGuangzhouChina
| | - Hong Quan
- Department of OrthopedicsShaoguan First People's Hospital Affiliated to Southern Medical UniversityShaoguanChina
| | - Shifeng Liu
- Orthopedics CenterDongguan Eighth People's HospitalDongguanChina
| | - Liqi Wu
- Department of OrthopedicsShaoguan First People's Hospital Affiliated to Southern Medical UniversityShaoguanChina
| | - Penghuan Wu
- Department of OrthopedicsShaoguan First People's Hospital Affiliated to Southern Medical UniversityShaoguanChina
- Orthopedics CenterZhujiang Hospital of Southern Medical UniversityGuangzhouChina
| | - Wenxing Lan
- Department of OrthopedicsShaoguan First People's Hospital Affiliated to Southern Medical UniversityShaoguanChina
| | - Yongjun Shao
- Department of OrthopedicsShaoguan First People's Hospital Affiliated to Southern Medical UniversityShaoguanChina
| | - Haomiao Li
- Orthopedics CenterThe Third Affiliated Hospital of Southern Medical UniversityOrthopedics institute of Guangdong ProvinceGuangzhouChina
| | - Kebing Chen
- Orthopedics CenterThe Third Affiliated Hospital of Southern Medical UniversityOrthopedics institute of Guangdong ProvinceGuangzhouChina
| | - Zhengbo Hu
- Department of OrthopedicsShaoguan First People's Hospital Affiliated to Southern Medical UniversityShaoguanChina
| |
Collapse
|
143
|
Nosacka RL, Delitto AE, Delitto D, Patel R, Judge SM, Trevino JG, Judge AR. Distinct cachexia profiles in response to human pancreatic tumours in mouse limb and respiratory muscle. J Cachexia Sarcopenia Muscle 2020; 11:820-837. [PMID: 32039571 PMCID: PMC7296265 DOI: 10.1002/jcsm.12550] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/20/2019] [Accepted: 01/07/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cancer cachexia is a life-threatening metabolic syndrome that causes significant loss of skeletal muscle mass and significantly increases mortality in cancer patients. Currently, there is an urgent need for better understanding of the molecular pathophysiology of this disease so that effective therapies can be developed. The majority of pre-clinical studies evaluating skeletal muscle's response to cancer have focused on one or two pre-clinical models, and almost all have focused specifically on limb muscles. In the current study, we reveal key differences in the histology and transcriptomic signatures of a limb muscle and a respiratory muscle in orthotopic pancreatic cancer patient-derived xenograft (PDX) mice. METHODS To create four cohorts of PDX mice evaluated in this study, tumours resected from four pancreatic ductal adenocarcinoma patients were portioned and attached to the pancreas of immunodeficient NSG mice. RESULTS Body weight, muscle mass, and fat mass were significantly decreased in each PDX line. Histological assessment of cryosections taken from the tibialis anterior (TA) and diaphragm (DIA) revealed differential effects of tumour burden on their morphology. Subsequent genome-wide microarray analysis on TA and DIA also revealed key differences between their transcriptomes in response to cancer. Genes up-regulated in the DIA were enriched for extracellular matrix protein-encoding genes and genes related to the inflammatory response, while down-regulated genes were enriched for mitochondria related protein-encoding genes. Conversely, the TA showed up-regulation of canonical atrophy-associated pathways such as ubiquitin-mediated protein degradation and apoptosis, and down-regulation of genes encoding extracellular matrix proteins. CONCLUSIONS These data suggest that distinct biological processes may account for wasting in different skeletal muscles in response to the same tumour burden. Further investigation into these differences will be critical for the future development of effective clinical strategies to counter cancer cachexia.
Collapse
Affiliation(s)
- Rachel L Nosacka
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, USA
| | - Andrea E Delitto
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, USA
| | - Dan Delitto
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, USA
| | - Rohan Patel
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, USA
| | - Sarah M Judge
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, USA
| | - Jose G Trevino
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, USA
| | - Andrew R Judge
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, USA
| |
Collapse
|
144
|
Garcia PL, Miller AL, Yoon KJ. Patient-Derived Xenograft Models of Pancreatic Cancer: Overview and Comparison with Other Types of Models. Cancers (Basel) 2020; 12:E1327. [PMID: 32456018 PMCID: PMC7281668 DOI: 10.3390/cancers12051327] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/11/2020] [Accepted: 05/19/2020] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer (PC) is anticipated to be second only to lung cancer as the leading cause of cancer-related deaths in the United States by 2030. Surgery remains the only potentially curative treatment for patients with pancreatic ductal adenocarcinoma (PDAC), the most common form of PC. Multiple recent preclinical studies focus on identifying effective treatments for PDAC, but the models available for these studies often fail to reproduce the heterogeneity of this tumor type. Data generated with such models are of unknown clinical relevance. Patient-derived xenograft (PDX) models offer several advantages over human cell line-based in vitro and in vivo models and models of non-human origin. PDX models retain genetic characteristics of the human tumor specimens from which they were derived, have intact stromal components, and are more predictive of patient response than traditional models. This review briefly describes the advantages and disadvantages of 2D cultures, organoids and genetically engineered mouse (GEM) models of PDAC, and focuses on the applications, characteristics, advantages, limitations, and the future potential of PDX models for improving the management of PDAC.
Collapse
Affiliation(s)
| | | | - Karina J. Yoon
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (P.L.G.); (A.L.M.)
| |
Collapse
|
145
|
Bhattacharyya R, Ha MJ, Liu Q, Akbani R, Liang H, Baladandayuthapani V. Personalized Network Modeling of the Pan-Cancer Patient and Cell Line Interactome. JCO Clin Cancer Inform 2020; 4:399-411. [PMID: 32374631 PMCID: PMC7265783 DOI: 10.1200/cci.19.00140] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2020] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Personalized network inference on diverse clinical and in vitro model systems across cancer types can be used to delineate specific regulatory mechanisms, uncover drug targets and pathways, and develop individualized predictive models in cancer. METHODS We developed TransPRECISE (personalized cancer-specific integrated network estimation model), a multiscale Bayesian network modeling framework, to analyze the pan-cancer patient and cell line interactome to identify differential and conserved intrapathway activities, to globally assess cell lines as representative models for patients, and to develop drug sensitivity prediction models. We assessed pan-cancer pathway activities for a large cohort of patient samples (> 7,700) from the Cancer Proteome Atlas across ≥ 30 tumor types, a set of 640 cancer cell lines from the MD Anderson Cell Lines Project spanning 16 lineages, and ≥ 250 cell lines' response to > 400 drugs. RESULTS TransPRECISE captured differential and conserved proteomic network topologies and pathway circuitry between multiple patient and cell line lineages: ovarian and kidney cancers shared high levels of connectivity in the hormone receptor and receptor tyrosine kinase pathways, respectively, between the two model systems. Our tumor stratification approach found distinct clinical subtypes of the patients represented by different sets of cell lines: patients with head and neck tumors were classified into two different subtypes that are represented by head and neck and esophagus cell lines and had different prognostic patterns (456 v 654 days of median overall survival; P = .02). High predictive accuracy was observed for drug sensitivities in cell lines across multiple drugs (median area under the receiver operating characteristic curve > 0.8) using Bayesian additive regression tree models with TransPRECISE pathway scores. CONCLUSION Our study provides a generalizable analytic framework to assess the translational potential of preclinical model systems and to guide pathway-based personalized medical decision making, integrating genomic and molecular data across model systems.
Collapse
Affiliation(s)
| | - Min Jin Ha
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Qingzhi Liu
- Department of Biostatistics, University of Michigan, Ann Arbor, MI
| | - Rehan Akbani
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
146
|
De Thaye E, Van de Vijver K, Van der Meulen J, Taminau J, Wagemans G, Denys H, Van Dorpe J, Berx G, Ceelen W, Van Bocxlaer J, De Wever O. Establishment and characterization of a cell line and patient-derived xenograft (PDX) from peritoneal metastasis of low-grade serous ovarian carcinoma. Sci Rep 2020; 10:6688. [PMID: 32317693 PMCID: PMC7174384 DOI: 10.1038/s41598-020-63738-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 03/13/2020] [Indexed: 12/24/2022] Open
Abstract
Peritoneal spread indicates poor prognosis in patients with serous ovarian carcinoma (SOC) and is generally treated by surgical cytoreduction and chemotherapy. Novel treatment options are urgently needed to improve patient outcome. Clinically relevant cell lines and patient-derived xenograft (PDX) models are of critical importance to therapeutic regimen evaluation. Here, a PDX model was established, by orthotopic engraftment after subperitoneal tumor slurry injection of low-grade SOC, resulting in an early-stage transplantable peritoneal metastasis (PM)-PDX model. Histology confirmed the micropapillary and cribriform growth pattern with intraluminal tumor budding and positivity for PAX8 and WT1. PM-PDX dissociated cells show an epithelial morphotype with a 42 h doubling time and 40% colony forming efficiency, they are low sensitive to platinum derivatives and highly sensitive to paclitaxel (IC50: 6.3 ± 2.2 nM, mean ± SEM). The patient primary tumor, PM, PM-PDX and derived cell line all show a KRAS c.35 G > T (p.(Gly12Val)) mutation and show sensitivity to the MEK inhibitor trametinib in vitro (IC50: 7.2 ± 0.5 nM, mean ± SEM) and in the PM mouse model. These preclinical models closely reflecting patient tumors are useful to further elucidate LGSOC disease progression, therapy response and resistance mechanisms.
Collapse
Affiliation(s)
- Elien De Thaye
- Laboratory of Medical Biochemistry and Clinical Analysis, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Koen Van de Vijver
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Joni Van der Meulen
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Molecular Diagnostics Ghent University Hospital, Ghent, Belgium
| | - Joachim Taminau
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Molecular and Cellular Oncology lab, Ghent University, Ghent, Belgium
| | - Glenn Wagemans
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Laboratory of Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Hannelore Denys
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Medical Oncology, Ghent University Hospital, Ghent, Belgium
| | - Jo Van Dorpe
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Geert Berx
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Molecular and Cellular Oncology lab, Ghent University, Ghent, Belgium
| | - Wim Ceelen
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of GI Surgery, Ghent University Hospital, Ghent, Belgium
| | - Jan Van Bocxlaer
- Laboratory of Medical Biochemistry and Clinical Analysis, Ghent University, Ghent, Belgium
| | - Olivier De Wever
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
- Laboratory of Experimental Cancer Research, Ghent University, Ghent, Belgium.
| |
Collapse
|
147
|
Kang HN, Kim JH, Park AY, Choi JW, Lim SM, Kim J, Shin EJ, Hong MH, Pyo KH, Yun MR, Kim DH, Lee H, Yoon SO, Kim DH, Park YM, Byeon HK, Jung I, Paik S, Koh YW, Cho BC, Kim HR. Establishment and characterization of patient-derived xenografts as paraclinical models for head and neck cancer. BMC Cancer 2020; 20:316. [PMID: 32293356 PMCID: PMC7160896 DOI: 10.1186/s12885-020-06786-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/25/2020] [Indexed: 02/08/2023] Open
Abstract
Background We investigated whether head and neck squamous cell carcinoma (HNSCC) patient-derived xenografts (PDXs) reaffirm patient responses to anti-cancer therapeutics. Methods Tumors from HNSCC patients were transplanted into immunodeficient mice and propagated via subsequent implantation. We evaluated established PDXs by histology, genomic profiling, and in vivo anti-cancer efficacy testing to confirm them as the authentic in vivo platform. Results From 62 HNSCCs, 15 (24%) PDXs were established. The primary cancer types were tongue (8), oropharynx (3), hypopharynx (1), ethmoid sinus cancer (1), supraglottic cancer (1), and parotid gland (1); six PDXs (40%) were established from biopsy specimens from advanced HNSCC. PDXs mostly retained donor characteristics and remained stable across passages. PIK3CA (H1047R), HRAS (G12D), and TP53 mutations (H193R, I195T, R248W, R273H, E298X) and EGFR, CCND1, MYC, and PIK3CA amplifications were identified. Using the acquisition method, biopsy showed a significantly higher engraftment rate when compared with that of surgical resection (100% [6/6] vs. 16.1% [9/56], P < 0.001). Specimens obtained from metastatic sites showed a significantly higher engraftment rate than did those from primary sites (100% [9/9] vs. 11.3% [6/53], P < 0.001). Three PDX models from HPV-positive tumors were established, as compared to 12 from HPV-negative (15.8% [3/19] and 27.9% [12/43] respectively, P = 0.311), suggesting that HPV positivity tends to show a low engraftment rate. Drug responses in PDX recapitulated the clinical responses of the matching patients with pan-HER inhibitors and pan-PI3K inhibitor. Conclusions Genetically and clinically annotated HNSCC PDXs could be useful preclinical tools for evaluating biomarkers, therapeutic targets, and new drug discovery.
Collapse
Affiliation(s)
- Han Na Kang
- JE-UK Institute for Cancer Research, JEUK Co. Ltd., Gumi-City, Kyungbuk, South Korea
| | - Jae-Hwan Kim
- JE-UK Institute for Cancer Research, JEUK Co. Ltd., Gumi-City, Kyungbuk, South Korea
| | - A-Young Park
- JE-UK Institute for Cancer Research, JEUK Co. Ltd., Gumi-City, Kyungbuk, South Korea
| | - Jae Woo Choi
- JE-UK Institute for Cancer Research, JEUK Co. Ltd., Gumi-City, Kyungbuk, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, Seongnam-si, South Korea
| | - Jinna Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun Joo Shin
- JE-UK Institute for Cancer Research, JEUK Co. Ltd., Gumi-City, Kyungbuk, South Korea
| | - Min Hee Hong
- Yonsei Cancer Center, Division of Medical Oncology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Kyoung-Ho Pyo
- JE-UK Institute for Cancer Research, JEUK Co. Ltd., Gumi-City, Kyungbuk, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Mi Ran Yun
- JE-UK Institute for Cancer Research, JEUK Co. Ltd., Gumi-City, Kyungbuk, South Korea
| | - Dong Hwi Kim
- JE-UK Institute for Cancer Research, JEUK Co. Ltd., Gumi-City, Kyungbuk, South Korea
| | - Hanna Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Sun Och Yoon
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Da Hee Kim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Young Min Park
- Department of Otorhinolaryngology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Hyung Kwon Byeon
- Department of Otolaryngology-Head and Neck Surgery Korea, University College of Medicine, Seoul, South Korea
| | - Inkyung Jung
- Department of Biostatistics and Medical Informatics, Yonsei University College of Medicine, Seoul, South Korea
| | - Soonmyung Paik
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Yoon Woo Koh
- Department of Otorhinolaryngology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, South Korea.
| | - Byoung Chul Cho
- JE-UK Institute for Cancer Research, JEUK Co. Ltd., Gumi-City, Kyungbuk, South Korea. .,Yonsei Cancer Center, Division of Medical Oncology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, South Korea.
| | - Hye Ryun Kim
- Yonsei Cancer Center, Division of Medical Oncology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, South Korea.
| |
Collapse
|
148
|
Abstract
Uveal melanoma (UM) is the most common primary intraocular malignancy in adults. UMs are usually initiated by a mutation in GNAQ or GNA11, unlike cutaneous melanomas, which usually harbour a BRAF or NRAS mutation. The annual incidence in Europe and the USA is ~6 per million population per year. Risk factors include fair skin, light-coloured eyes, congenital ocular melanocytosis, ocular melanocytoma and the BAP1-tumour predisposition syndrome. Ocular treatment aims at preserving the eye and useful vision and, if possible, preventing metastases. Enucleation has largely been superseded by various forms of radiotherapy, phototherapy and local tumour resection, often administered in combination. Ocular outcomes are best with small tumours not extending close to the optic disc and/or fovea. Almost 50% of patients develop metastatic disease, which usually involves the liver, and is usually fatal within 1 year. Although UM metastases are less responsive than cutaneous melanoma to chemotherapy or immune checkpoint inhibitors, encouraging results have been reported with partial hepatectomy for solitary metastases, with percutaneous hepatic perfusion with melphalan or with tebentafusp. Better insight into tumour immunology and metabolism may lead to new treatments.
Collapse
|
149
|
Meneceur S, Linge A, Meinhardt M, Hering S, Löck S, Bütof R, Krex D, Schackert G, Temme A, Baumann M, Krause M, von Neubeck C. Establishment and Characterisation of Heterotopic Patient-Derived Xenografts for Glioblastoma. Cancers (Basel) 2020; 12:cancers12040871. [PMID: 32260145 PMCID: PMC7226316 DOI: 10.3390/cancers12040871] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/27/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is an aggressive brain tumour with a patient median survival of approximately 14 months. The development of innovative treatment strategies to increase the life span and quality of life of patients is hence essential. This requires the use of appropriate glioblastoma models for preclinical testing, which faithfully reflect human cancers. The aim of this study was to establish glioblastoma patient-derived xenografts (PDXs) by heterotopic transplantation of tumour pieces in the axillae of NMRI nude mice. Ten out of 22 patients' samples gave rise to tumours in mice. Their human origin was confirmed by microsatellite analyses, though minor changes were observed. The glioblastoma nature of the PDXs was corroborated by pathological evaluation. Latency times spanned from 48.5 to 370.5 days in the first generation. Growth curve analyses revealed an increase in the growth rate with increasing passages. The methylation status of the MGMT promoter in the primary material was maintained in the PDXs. However, a trend towards a more methylated pattern could be found. A correlation was observed between the take in mice and the proportion of Sox2+ cells (r = 0.49, p = 0.016) and nestin+ cells (r = 0.55, p = 0.007). Our results show that many PDXs maintain key features of the patients' samples they derive from. They could thus be used as preclinical models to test new therapies and biomarkers.
Collapse
Affiliation(s)
- Sarah Meneceur
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz- Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (A.L.); (S.L.); (R.B.); (M.B.); (M.K.); (C.v.N.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology–OncoRay, 01307 Dresden, Germany
- Correspondence:
| | - Annett Linge
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz- Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (A.L.); (S.L.); (R.B.); (M.B.); (M.K.); (C.v.N.)
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany; (G.S.); (A.T.)
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumour Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| | - Matthias Meinhardt
- Institute for Pathology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany;
| | - Sandra Hering
- Institute for Legal Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany;
| | - Steffen Löck
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz- Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (A.L.); (S.L.); (R.B.); (M.B.); (M.K.); (C.v.N.)
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany; (G.S.); (A.T.)
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Rebecca Bütof
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz- Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (A.L.); (S.L.); (R.B.); (M.B.); (M.K.); (C.v.N.)
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumour Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| | - Dietmar Krex
- Department of Neurosurgery, Medical Faculty and University Hospital Carl Gustav Carus, 01307 Dresden, Germany;
| | - Gabriele Schackert
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany; (G.S.); (A.T.)
- National Center for Tumour Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
- Department of Neurosurgery, Medical Faculty and University Hospital Carl Gustav Carus, 01307 Dresden, Germany;
| | - Achim Temme
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany; (G.S.); (A.T.)
- National Center for Tumour Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
- Department of Neurosurgery, Medical Faculty and University Hospital Carl Gustav Carus, 01307 Dresden, Germany;
| | - Michael Baumann
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz- Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (A.L.); (S.L.); (R.B.); (M.B.); (M.K.); (C.v.N.)
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Mechthild Krause
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz- Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (A.L.); (S.L.); (R.B.); (M.B.); (M.K.); (C.v.N.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology–OncoRay, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany; (G.S.); (A.T.)
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumour Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| | - Cläre von Neubeck
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz- Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (A.L.); (S.L.); (R.B.); (M.B.); (M.K.); (C.v.N.)
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany; (G.S.); (A.T.)
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Particle Therapy, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| |
Collapse
|
150
|
Leiting JL, Murphy SJ, Bergquist JR, Hernandez MC, Ivanics T, Abdelrahman AM, Yang L, Lynch I, Smadbeck JB, Cleary SP, Nagorney DM, Torbenson MS, Graham RP, Roberts LR, Gores GJ, Smoot RL, Truty MJ. Biliary tract cancer patient-derived xenografts: Surgeon impact on individualized medicine. JHEP Rep 2020; 2:100068. [PMID: 32181445 PMCID: PMC7066236 DOI: 10.1016/j.jhepr.2020.100068] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 01/08/2020] [Accepted: 01/08/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND & AIMS Biliary tract tumors are uncommon but highly aggressive malignancies with poor survival outcomes. Due to their low incidence, research into effective therapeutics has been limited. Novel research platforms for pre-clinical studies are desperately needed. We sought to develop a patient-derived biliary tract cancer xenograft catalog. METHODS With appropriate consent and approval, surplus malignant tissues were obtained from surgical resection or radiographic biopsy and implanted into immunocompromised mice. Mice were monitored for xenograft growth. Established xenografts were verified by a hepatobiliary pathologist. Xenograft characteristics were correlated with original patient/tumor characteristics and oncologic outcomes. A subset of xenografts were then genomically characterized using Mate Pair sequencing (MPseq). RESULTS Between October 2013 and January 2018, 87 patients with histologically confirmed biliary tract carcinomas were enrolled. Of the 87 patients, 47 validated PDX models were successfully generated. The majority of the PDX models were created from surgical resection specimens (n = 44, 94%), which were more likely to successfully engraft when compared to radiologic biopsies (p = 0.03). Histologic recapitulation of original patient tumor morphology was observed in all xenografts. Successful engraftment was an independent predictor for worse recurrence-free survival. MPseq showed genetically diverse tumors with frequent alterations of CDKN2A, SMAD4, NRG1, TP53. Sequencing also identified worse survival in patients with tumors containing tetraploid genomes. CONCLUSIONS This is the largest series of biliary tract cancer xenografts reported to date. Histologic and genomic analysis of patient-derived xenografts demonstrates accurate recapitulation of original tumor morphology with direct correlations to patient outcomes. Successful development of biliary cancer tumografts is feasible and may be used to direct subsequent therapy in high recurrence risk patients. LAY SUMMARY Patient biliary tract tumors grown in immunocompromised mice are an invaluable resource in the treatment of biliary tract cancers. They can be used to guide individualized cancer treatment in high-risk patients.
Collapse
Key Words
- CCA, cholangiocarcinoma
- ECM, extracellular matrix
- GBCA, gallbladder carcinoma
- HRs, hazard ratios
- LOH, loss of heterozygosity
- MatePair sequencing
- OPTR, overall patient take rate
- OS, overall survival
- PDX, patient-derived xenograft
- Patient-derived xenografts
- TTF, time to tumor formation
- TTH, time to tumor harvest
- biliary tract
- cholangiocarcinoma
- dCCA, distal cholangiocarcinoma
- gallbladder carcinoma
- iCCA, intrahepatic cholangiocarcinoma
- pCCA, perihilar cholangiocarcinoma
Collapse
Affiliation(s)
| | | | | | | | - Tommy Ivanics
- Department of Surgery, Henry Ford Medical Center, Detroit, MI
| | | | - Lin Yang
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Isaac Lynch
- Department of Surgery, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | - Lewis R. Roberts
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Gregory J. Gores
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | | | | |
Collapse
|