201
|
Mihara H, Kugawa M, Sayo K, Tao F, Shinohara M, Nishikawa M, Sakai Y, Akama T, Kojima N. Improved Oxygen Supply to Multicellular Spheroids Using A Gas-permeable Plate and Embedded Hydrogel Beads. Cells 2019; 8:cells8060525. [PMID: 31159231 PMCID: PMC6627619 DOI: 10.3390/cells8060525] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/18/2019] [Accepted: 05/25/2019] [Indexed: 12/12/2022] Open
Abstract
Culture systems for three-dimensional tissues, such as multicellular spheroids, are indispensable for high-throughput screening of primary or patient-derived xenograft (PDX)-expanded cancer tissues. Oxygen supply to the center of such spheroids is particularly critical for maintaining cellular functions as well as avoiding the development of a necrotic core. In this study, we evaluated two methods to enhance oxygen supply: (1) using a culture plate with a gas-permeable polydimethylsiloxane (PDMS) membrane on the bottom, and; (2) embedding hydrogel beads in the spheroids. Culturing spheroids on PDMS increased cell growth and affected glucose/lactate metabolism and CYP3A4 mRNA expression and subsequent enzyme activity. The spheroids, comprised of 5000 Hep G2 cells and 5000 20 µm-diameter hydrogel beads, did not develop a necrotic core for nine days when cultured on a gas-permeable sheet. In contrast, central necrosis in spheroids lacking hydrogel beads was observed after day 3 of culture, even when using PDMS. These results indicate that the combination of gas-permeable culture equipment and embedded hydrogel beads improves culture 3D spheroids produced from primary or PDX-expanded tumor cells.
Collapse
Affiliation(s)
- Hirotaka Mihara
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan.
| | - Mai Kugawa
- Faculty of Science, International College of Arts and Science, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan.
| | - Kanae Sayo
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan.
| | - Fumiya Tao
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan.
| | - Marie Shinohara
- Department of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Masaki Nishikawa
- Department of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Yasuyuki Sakai
- Department of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Takeshi Akama
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan.
| | - Nobuhiko Kojima
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan.
- Faculty of Science, International College of Arts and Science, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan.
| |
Collapse
|
202
|
Mer AS, Ba-Alawi W, Smirnov P, Wang YX, Brew B, Ortmann J, Tsao MS, Cescon DW, Goldenberg A, Haibe-Kains B. Integrative Pharmacogenomics Analysis of Patient-Derived Xenografts. Cancer Res 2019; 79:4539-4550. [DOI: 10.1158/0008-5472.can-19-0349] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/16/2019] [Accepted: 05/23/2019] [Indexed: 11/16/2022]
|
203
|
Lin HS, Huang YL, Wang YRS, Hsiao E, Hsu TA, Shiao HY, Jiaang WT, Sampurna BP, Lin KH, Wu MS, Lai GM, Yuh CH. Identification of Novel Anti-Liver Cancer Small Molecules with Better Therapeutic Index than Sorafenib via Zebrafish Drug Screening Platform. Cancers (Basel) 2019; 11:cancers11060739. [PMID: 31141996 PMCID: PMC6628114 DOI: 10.3390/cancers11060739] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 05/21/2019] [Accepted: 05/26/2019] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) ranks as the fourth leading cause of cancer-related deaths worldwide. Sorafenib was the only U.S. Food and Drug Administration (FDA) approved drug for treating advanced HCC until recently, so development of new target therapy is urgently needed. In this study, we established a zebrafish drug screening platform and compared the therapeutic effects of two multiple tyrosine kinase inhibitors, 419S1 and 420S1, with Sorafenib. All three compounds exhibited anti-angiogenesis abilities in immersed fli1:EGFP transgenic embryos and the half inhibition concentration (IC50) was determined. 419S1 exhibited lower hepatoxicity and embryonic toxicity than 420S1 and Sorafenib, and the half lethal concentration (LC50) was determined. The therapeutic index (LC50/IC50) for 419S1 was much higher than for Sorafenib and 420S1. The compounds were either injected retro-orbitally or by oral gavage to adult transgenic zebrafish with HCC. The compounds not only rescued the pathological feature, but also reversed the expression levels of cell-cycle-related genes and protein levels of a proliferation marker. Using a patient-derived-xenograft assay, we found that the effectiveness of 419S1 and 420S1 in preventing liver cancer proliferation is better than that of Sorafenib. With integrated efforts and the advantage of the zebrafish platform, we can find more effective and safe drugs for HCC treatment and screen for personalized medicine.
Collapse
Affiliation(s)
- Han-Syuan Lin
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan.
| | - Yi-Luen Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan.
| | - Yi-Rui Stefanie Wang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan.
| | - Eugene Hsiao
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan.
| | - Tsu-An Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan.
| | - Hui-Yi Shiao
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan.
| | - Weir-Torn Jiaang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan.
| | - Bonifasius Putera Sampurna
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan.
| | - Kuan-Hao Lin
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan.
| | - Ming-Shun Wu
- Division of Gastroenterology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan.
| | - Gi-Ming Lai
- TMU Research Center of Cancer Translational Medicine, Taipei Municipal Wanfang Hospital, Taipei 11696, Taiwan.
| | - Chiou-Hwa Yuh
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan.
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30010, Taiwan.
- Institute of Bioinformatics and Structural Biology, National Tsing-Hua University, Hsinchu 30013, Taiwan.
- Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
204
|
Blumer T, Fofana I, Matter MS, Wang X, Montazeri H, Calabrese D, Coto-Llerena M, Boldanova T, Nuciforo S, Kancherla V, Tornillo L, Piscuoglio S, Wieland S, Terracciano LM, Ng CKY, Heim MH. Hepatocellular Carcinoma Xenografts Established From Needle Biopsies Preserve the Characteristics of the Originating Tumors. Hepatol Commun 2019; 3:971-986. [PMID: 31334445 PMCID: PMC6601318 DOI: 10.1002/hep4.1365] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/19/2019] [Indexed: 12/26/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer‐related deaths worldwide. Treatment options for patients with advanced‐stage disease are limited. A major obstacle in drug development is the lack of an in vivo model that accurately reflects the broad spectrum of human HCC. Patient‐derived xenograft (PDX) tumor mouse models could overcome the limitations of cancer cell lines. PDX tumors maintain the genetic and histologic heterogeneity of the originating tumors and are used for preclinical drug development in various cancers. Controversy exists about their genetic and molecular stability through serial passaging in mice. We aimed to establish PDX models from human HCC biopsies and to characterize their histologic and molecular stability during serial passaging. A total of 54 human HCC needle biopsies that were derived from patients with various underlying liver diseases and tumor stages were transplanted subcutaneously into immunodeficient, nonobese, diabetic/severe combined immunodeficiency gamma‐c mice; 11 successfully engrafted. All successfully transplanted HCCs were Edmondson grade III or IV. HCC PDX tumors retained the histopathologic, transcriptomic, and genomic characteristics of the original HCC biopsies over 6 generations of retransplantation. These characteristics included Edmondson grade, expression of tumor markers, tumor gene signature, tumor‐associated mutations, and copy number alterations. Conclusion: PDX mouse models can be established from undifferentiated HCCs, with an overall success rate of approximately 20%. The transplanted tumors represent the entire spectrum of the molecular landscape of HCCs and preserve the characteristics of the originating tumors through serial passaging. HCC PDX models are a promising tool for preclinical personalized drug development.
Collapse
Affiliation(s)
- Tanja Blumer
- Department of Biomedicine University Hospital Basel, University of Basel Basel Switzerland
| | - Isabel Fofana
- Department of Biomedicine University Hospital Basel, University of Basel Basel Switzerland
| | - Matthias S Matter
- Institute of Pathology University Hospital Basel, University of Basel Basel Switzerland
| | - Xueya Wang
- Department of Biomedicine University Hospital Basel, University of Basel Basel Switzerland
| | - Hesam Montazeri
- Institute of Pathology University Hospital Basel, University of Basel Basel Switzerland.,Department of Bioinformatics, Institute of Biochemistry and Biophysics University of Tehran Tehran Iran
| | - Diego Calabrese
- Department of Biomedicine University Hospital Basel, University of Basel Basel Switzerland
| | - Mairene Coto-Llerena
- Department of Biomedicine University Hospital Basel, University of Basel Basel Switzerland
| | - Tujana Boldanova
- Department of Biomedicine University Hospital Basel, University of Basel Basel Switzerland.,Division of Gastroenterology and Hepatology Clarunis, University Hospital Basel, University of Basel Basel Switzerland
| | - Sandro Nuciforo
- Department of Biomedicine University Hospital Basel, University of Basel Basel Switzerland
| | - Venkatesh Kancherla
- Institute of Pathology University Hospital Basel, University of Basel Basel Switzerland
| | - Luigi Tornillo
- Institute of Pathology University Hospital Basel, University of Basel Basel Switzerland
| | - Salvatore Piscuoglio
- Institute of Pathology University Hospital Basel, University of Basel Basel Switzerland.,Visceral Surgery Research Laboratory, Clarunis, Department of Biomedicine University of Basel Basel Switzerland
| | - Stefan Wieland
- Department of Biomedicine University Hospital Basel, University of Basel Basel Switzerland
| | - Luigi M Terracciano
- Institute of Pathology University Hospital Basel, University of Basel Basel Switzerland
| | - Charlotte K Y Ng
- Department of Biomedicine University Hospital Basel, University of Basel Basel Switzerland.,Institute of Pathology University Hospital Basel, University of Basel Basel Switzerland.,Department for Biomedical Research University of Bern Bern Switzerland
| | - Markus H Heim
- Department of Biomedicine University Hospital Basel, University of Basel Basel Switzerland.,Division of Gastroenterology and Hepatology Clarunis, University Hospital Basel, University of Basel Basel Switzerland
| |
Collapse
|
205
|
Cho JH, Kim JS, Kim ST, Hong JY, Park JO, Park YS, Nam DH, Lee DW, Lee J. Selective colony area method for heterogeneous patient-derived tumor cell lines in anti-cancer drug screening system. PLoS One 2019; 14:e0215080. [PMID: 30995234 PMCID: PMC6469764 DOI: 10.1371/journal.pone.0215080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 03/26/2019] [Indexed: 12/20/2022] Open
Abstract
We aimed to establish a fluorescence intensity-based colony area sweeping method by selecting the area of highest viability among patient-derived cancer cells (PDC) which has high tumor heterogeneity. Five gastric cancer cell lines and PDCs were screened with 24 small molecule compounds using a 3D micropillar/microwell chip. 100 tumor cells per well were immobilized in alginate, treated with the compounds, and then stained and scanned for viable cells. Dose response curves and IC50 values were obtained based on total or selected area intensity based on fluorescence. Unlike homogeneous cell lines, PDC comprised of debris and low-viability cells, which resulted in an inaccurate estimation of cell viability using total fluorescence intensity as determined by high IC50 values. However, the IC50 of these cells was lower and accurate when calculated based on the selected-colony-area method that eliminated the intensity offset associated with the heterogeneous nature of PDC. The selected-colony-area method was optimized to accurately predict drug response in micropillar environment using heterogeneous nature of PDCs.
Collapse
Affiliation(s)
- Jang Ho Cho
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ju-Sun Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seung Tae Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jung Yong Hong
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joon Oh Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Suk Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Do-Hyun Nam
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dong Woo Lee
- Department of Biomedical Engineering, Konyang University, Daejeon, Korea
- * E-mail: (DWL); (JL)
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- * E-mail: (DWL); (JL)
| |
Collapse
|
206
|
Rituximab Decreases Lymphoproliferative Tumor Formation in Hepatopancreaticobiliary and Gastrointestinal Cancer Patient-Derived Xenografts. Sci Rep 2019; 9:5901. [PMID: 30976061 PMCID: PMC6459856 DOI: 10.1038/s41598-019-42470-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 03/28/2019] [Indexed: 12/12/2022] Open
Abstract
High engraftment rates are critical to any patient-derived xenograft (PDX) program and the loss of PDX models due to the development of lymphoproliferative tumors (LTs) is costly and inefficient. We hypothesized that routine injection of rituximab, an anti-CD20 antibody, at the time of implantation would reduce the incidence of LTs. Rituximab injection was added to the standard PDX engraftment protocol. Univariate analysis and multivariate logistic regression were used to determine the significance of various factors. A total of 811 generations of PDX were implanted with 406 receiving rituximab with implantation. On multivariable analysis, rituximab was an independent factor for decreased LT formation across the entire cohort (OR 0.465, 95% CI 0.271–0.797, p = 0.005). Hepatocellular carcinomas (OR 0.319, 95% CI 0.107–0.949, p = 0.040) and cholangiocarcinomas (OR 0.185, 95% CI 0.049–0.696, p = 0.113) were the specific malignant histologic subtypes that demonstrated the greatest benefit. The frequency of LTs decreased across the entire cohort with rituximab administration and PDX tumors that are traditionally associated with higher rates of LT formation, HCCs and CCAs, appear to benefit the most from rituximab treatment. Routine use of rituximab at the time of tumor implantation may have significant programmatic benefits for laboratories that utilize PDX models.
Collapse
|
207
|
Buhl IK, Jensen PB, Kappel Buhl AS, Knudsen S. A drug response predictor to guide treatment for breast cancer. Pharmacogenomics 2019; 20:307-309. [DOI: 10.2217/pgs-2018-0195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Ida Kappel Buhl
- Oncology Venture, Hørsholm, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|
208
|
Gioeli D, Snow CJ, Simmers MB, Hoang SA, Figler RA, Allende JA, Roller DG, Parsons JT, Wulfkuhle JD, Petricoin EF, Bauer TW, Wamhoff BR. Development of a multicellular pancreatic tumor microenvironment system using patient-derived tumor cells. LAB ON A CHIP 2019; 19:1193-1204. [PMID: 30839006 PMCID: PMC7486791 DOI: 10.1039/c8lc00755a] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The development of drugs to treat cancer is hampered by the inefficiency of translating pre-clinical in vitro monoculture and mouse studies into clinical benefit. There is a critical need to improve the accuracy of evaluating pre-clinical drug efficacy through the development of more physiologically relevant models. In this study, a human triculture 3D in vitro tumor microenvironment system (TMES) was engineered to accurately mimic the tumor microenvironment. The TMES recapitulates tumor hemodynamics and biological transport with co-cultured human microvascular endothelial cells, pancreatic ductal adenocarcinoma, and pancreatic stellate cells. We demonstrate that significant tumor cell transcriptomic changes occur in the TMES that correlate with the in vivo xenograft and patient transcriptome. Treatment with therapeutically relevant doses of chemotherapeutics yields responses paralleling the patients' clinical responses. Thus, this model provides a unique platform to rigorously evaluate novel therapies and is amenable to using patient tumor material directly, with applicability for patient avatars.
Collapse
Affiliation(s)
- Daniel Gioeli
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
- Cancer Center Member, University of Virginia, Charlottesville, Virginia
- HemoShear Therapeutics, Charlottesville, Virginia
| | | | | | | | | | - J. Ashe Allende
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Devin G. Roller
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - J. Thomas Parsons
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
- Cancer Center Member, University of Virginia, Charlottesville, Virginia
| | - Julia D. Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Emanuel F. Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Todd W. Bauer
- Department of Surgery, University of Virginia, Charlottesville, Virginia
- Cancer Center Member, University of Virginia, Charlottesville, Virginia
| | | |
Collapse
|
209
|
Liu Y, Wang Y, Cao Z, Gao Y. Changes in the Urinary Proteome in a Patient-Derived Xenograft (PDX) Nude Mouse Model of Colorectal Tumor. Sci Rep 2019; 9:4975. [PMID: 30899049 PMCID: PMC6428931 DOI: 10.1038/s41598-019-41361-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/07/2019] [Indexed: 02/06/2023] Open
Abstract
In this report, the urinary proteome from a patient-derived xenograft (PDX) model was examined at the peptide level to study the origins of urinary proteins in tumor-bearing nude mice. Urine was collected from PDX mice before and after colorectal tumor implantation. A total of 4,318 unique peptides were identified, and 78 unambiguous human-origin peptides were discerned in the PDX model urine. Unlike the differential urinary protein composition of tumor-bearing immunocompetent rat models, the differential urinary proteins in the PDX model did not include host immune-response proteins. This study demonstrates that tumor-secreted proteins can be observed in the urine proteome of the PDX model. However, immune-response proteins, which are very early candidate tumor biomarkers, are not present in the urine of PDX model mice; this absence is due to immune deficiency. Therefore, immunodeficient animals may not be suitable models for searching for early immunity-associated tumor biomarkers in the urine.
Collapse
Affiliation(s)
- Yongtao Liu
- Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Department of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Youzhu Wang
- Beijing Percans Oncology Co., Ltd, Beijing, China
| | - Zhixiang Cao
- Beijing Percans Oncology Co., Ltd, Beijing, China
| | - Youhe Gao
- Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Department of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University, Beijing, China.
| |
Collapse
|
210
|
Ward C, Meehan J, Gray M, Kunkler IH, Langdon SP, Murray A, Argyle D. Preclinical Organotypic Models for the Assessment of Novel Cancer Therapeutics and Treatment. Curr Top Microbiol Immunol 2019. [PMID: 30859401 DOI: 10.1007/82_2019_159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The immense costs in both financial terms and preclinical research effort that occur in the development of anticancer drugs are unfortunately not matched by a substantial increase in improved clinical therapies due to the high rate of failure during clinical trials. This may be due to issues with toxicity or lack of clinical effectiveness when the drug is evaluated in patients. Currently, much cancer research is driven by the need to develop therapies that can exploit cancer cell adaptations to conditions in the tumor microenvironment such as acidosis and hypoxia, the requirement for more-specific, targeted treatments, or the exploitation of 'precision medicine' that can target known genomic changes in patient DNA. The high attrition rate for novel anticancer therapies suggests that the preclinical methods used in screening anticancer drugs need improvement. This chapter considers the advantages and disadvantages of 3D organotypic models in both cancer research and cancer drug screening, particularly in the areas of targeted drugs and the exploitation of genomic changes that can be used for therapeutic advantage in precision medicine.
Collapse
Affiliation(s)
- Carol Ward
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Roslin, Midlothian, EH25 9RG, Edinburgh, UK.
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU, Edinburgh, UK.
| | - James Meehan
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU, Edinburgh, UK
- School of Engineering and Physical Sciences, Institute of Sensors, Signals and Systems, Heriot-Watt University, EH14 4AS, Edinburgh, UK
| | - Mark Gray
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Roslin, Midlothian, EH25 9RG, Edinburgh, UK
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU, Edinburgh, UK
| | - Ian H Kunkler
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU, Edinburgh, UK
| | - Simon P Langdon
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU, Edinburgh, UK
| | - Alan Murray
- School of Engineering, Faraday Building, The King's Buildings, Mayfield Road, EH9 3JL, Edinburgh, UK
| | - David Argyle
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Roslin, Midlothian, EH25 9RG, Edinburgh, UK
| |
Collapse
|
211
|
Johanna I, Straetemans T, Heijhuurs S, Aarts-Riemens T, Norell H, Bongiovanni L, de Bruin A, Sebestyen Z, Kuball J. Evaluating in vivo efficacy - toxicity profile of TEG001 in humanized mice xenografts against primary human AML disease and healthy hematopoietic cells. J Immunother Cancer 2019; 7:69. [PMID: 30871629 PMCID: PMC6419469 DOI: 10.1186/s40425-019-0558-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 03/04/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND γ9δ2T cells, which express Vγ9 and Vδ2 chains of the T cell receptor (TCR), mediate cancer immune surveillance by sensing early metabolic changes in malignant leukemic blast and not their healthy hematopoietic stem counterparts via the γ9δ2TCR targeting joined conformational and spatial changes of CD277 at the cell membrane (CD277J). This concept led to the development of next generation CAR-T cells, so-called TEGs: αβT cells Engineered to express a defined γδTCR. The high affinity γ9δ2TCR clone 5 has recently been selected within the TEG format as a clinical candidate (TEG001). However, exploring safety and efficacy against a target, which reflects an early metabolic change in tumor cells, remains challenging given the lack of appropriate tools. Therefore, we tested whether TEG001 is able to eliminate established leukemia in a primary disease model, without harming other parts of the healthy hematopoiesis in vivo. METHODS Separate sets of NSG mice were respectively injected with primary human acute myeloid leukemia (AML) blasts and cord blood-derived human progenitor cells from healthy donors. These mice were then treated with TEG001 and mock cells. Tumor burden and human cells engraftment were measured in peripheral blood and followed up over time by quantifying for absolute cell number by flow cytometry. Statistical analysis was performed using non-parametric 2-tailed Mann-Whitney t-test. RESULTS We successfully engrafted primary AML blasts and healthy hematopoietic cells after 6-8 weeks. Here we report that metabolic cancer targeting through TEG001 eradicated established primary leukemic blasts in vivo, while healthy hematopoietic compartments derived from human cord-blood remained unharmed in spite of TEGs persistence up to 50 days after infusion. No additional signs of off-target toxicity were observed in any other tissues. CONCLUSION Within the limitations of humanized PD-X models, targeting CD277J by TEG001 is safe and efficient. Therefore, we have initiated clinical testing of TEG001 in a phase I first-in-human clinical trial (NTR6541; date of registration 25 July 2017).
Collapse
Affiliation(s)
- Inez Johanna
- Department of Hematology and Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Trudy Straetemans
- Department of Hematology and Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sabine Heijhuurs
- Department of Hematology and Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tineke Aarts-Riemens
- Department of Hematology and Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Håkan Norell
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Laura Bongiovanni
- Department of Pathobiology, Faculty of Veterinary Medicine, Dutch Molecular Pathology Center, Utrecht University, Utrecht, The Netherlands
| | - Alain de Bruin
- Department of Pathobiology, Faculty of Veterinary Medicine, Dutch Molecular Pathology Center, Utrecht University, Utrecht, The Netherlands
| | - Zsolt Sebestyen
- Department of Hematology and Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jürgen Kuball
- Department of Hematology and Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
212
|
Mazzocchi A, Devarasetty M, Herberg S, Petty WJ, Marini F, Miller L, Kucera G, Dukes DK, Ruiz J, Skardal A, Soker S. Pleural Effusion Aspirate for use in 3D Lung Cancer Modeling and Chemotherapy Screening. ACS Biomater Sci Eng 2019; 5:1937-1943. [PMID: 31723594 DOI: 10.1021/acsbiomaterials.8b01356] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Lung cancer is the leading cause of cancer-related death worldwide yet in vitro disease models have been limited to traditional 2D culture utilizing cancer cell lines. In contrast, recently developed 3D models (organoids) have been adopted by researchers to improve the physiological relevance of laboratory study. We have hypothesized that 3D hydrogel-based models will allow for improved disease replication and characterization over standard 2D culture using cells taken directly from patients. Here, we have leveraged the use of 3D hydrogel-based models to create lung cancer organoids using a unique cell source, pleural effusion aspirate, from multiple lung cancer patients. With these 3D models, we have characterized the cell populations comprising the pleural effusion aspirate and have tracked phenotypic changes that develop during short-term in vitro culture. We found that isolated, patient cells placed directly into organoids created anatomically relevant structures and exhibited lung cancer specific behaviors. On the other hand, cells first grown in plastic dishes and then cultured in 3D did not create similar structures. Further, we have been able to compare chemotherapeutic response of patient cells between 2D and 3D cell culture systems. Our results show that cells in 2D culture were more sensitive to treatment when compared with 3D organoids. Collectively, we have been able to utilize tumor cells from pleural effusion fluid of lung cancer patients to create organoids that display in vivo like anatomy and drug response and thus could serve as more accurate disease models for study of tumor progression and drug development.
Collapse
Affiliation(s)
- Andrea Mazzocchi
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA.,Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Wake Forest School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Mahesh Devarasetty
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
| | - Samuel Herberg
- Current address: SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210
| | - William J Petty
- Department of Cancer Biology, Wake Forest School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.,Comprehensive Cancer Center at Wake Forest Baptist Medical, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Frank Marini
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA.,Department of Cancer Biology, Wake Forest School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Lance Miller
- Department of Cancer Biology, Wake Forest School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.,Comprehensive Cancer Center at Wake Forest Baptist Medical, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Gregory Kucera
- Department of Cancer Biology, Wake Forest School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.,Comprehensive Cancer Center at Wake Forest Baptist Medical, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - David K Dukes
- Current address: Alabama College of Osteopathic Medicine, 445 Health Sciences Blvd, Dothan, AL 36303, USA
| | - Jimmy Ruiz
- Department of Cancer Biology, Wake Forest School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.,Comprehensive Cancer Center at Wake Forest Baptist Medical, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Aleksander Skardal
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA.,Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Wake Forest School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.,Department of Cancer Biology, Wake Forest School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.,Comprehensive Cancer Center at Wake Forest Baptist Medical, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA.,Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Wake Forest School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.,Department of Cancer Biology, Wake Forest School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.,Comprehensive Cancer Center at Wake Forest Baptist Medical, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| |
Collapse
|
213
|
Parolia A, Venalainen E, Xue H, Mather R, Lin D, Wu R, Pucci P, Rogalski J, Evans JR, Feng F, Collins CC, Wang Y, Crea F. The long noncoding RNA HORAS5 mediates castration-resistant prostate cancer survival by activating the androgen receptor transcriptional program. Mol Oncol 2019; 13:1121-1136. [PMID: 30776192 PMCID: PMC6487714 DOI: 10.1002/1878-0261.12471] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 01/17/2019] [Accepted: 01/27/2019] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is driven by the androgen receptor (AR)‐signaling axis. Hormonal therapy often mitigates PCa progression, but a notable number of cases progress to castration‐resistant PCa (CRPC). CRPC retains AR activity and is incurable. Long noncoding RNA (lncRNA) represent an uncharted region of the transcriptome. Several lncRNA have been recently described to mediate oncogenic functions, suggesting that these molecules can be potential therapeutic targets. Here, we identified CRPC‐associated lncRNA by analyzing patient‐derived xenografts (PDXs) and clinical data. Subsequently, we characterized one of the CRPC‐promoting lncRNA,HORAS5, in vitro and in vivo. We demonstrated that HORAS5 is a stable, cytoplasmic lncRNA that promotes CRPC proliferation and survival by maintaining AR activity under androgen‐depleted conditions. Most strikingly, knockdown of HORAS5 causes a significant reduction in the expression of AR itself and oncogenic AR targets such as KIAA0101. Elevated expression of HORAS5 is also associated with worse clinical outcomes in patients. Our results from HORAS5 inhibition in in vivo models further confirm that HORAS5 is a viable therapeutic target for CRPC. Thus, we posit that HORAS5 is a novel, targetable mediator of CRPC through its essential role in the maintenance of oncogenic AR activity. Overall, this study adds to our mechanistic understanding of how lncRNA function in cancer progression.
Collapse
Affiliation(s)
- Abhijit Parolia
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Hui Xue
- British Columbia Cancer Research Centre, Vancouver, Canada.,Vancouver Prostate Centre, Canada
| | - Rebecca Mather
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Dong Lin
- British Columbia Cancer Research Centre, Vancouver, Canada.,Vancouver Prostate Centre, Canada
| | - Rebecca Wu
- British Columbia Cancer Research Centre, Vancouver, Canada
| | - Perla Pucci
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Jason Rogalski
- Proteomics Core Facility, Centre for High-Throughput Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
| | - Joseph R Evans
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Felix Feng
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | | | - Yuzhuo Wang
- British Columbia Cancer Research Centre, Vancouver, Canada.,Vancouver Prostate Centre, Canada
| | - Francesco Crea
- British Columbia Cancer Research Centre, Vancouver, Canada.,School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| |
Collapse
|
214
|
Dzobo K, Rowe A, Senthebane DA, AlMazyadi MAM, Patten V, Parker MI. Three-Dimensional Organoids in Cancer Research: The Search for the Holy Grail of Preclinical Cancer Modeling. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 22:733-748. [PMID: 30571609 DOI: 10.1089/omi.2018.0172] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Most solid tumors become therapy resistant and will relapse, with no durable treatment option available. One major impediment to our understanding of cancer biology and finding innovative approaches to cancer treatment stems from the lack of better preclinical tumor models that address and explain tumor heterogeneity and person-to-person differences in therapeutic and toxic responses. Past cancer research has been driven by inadequate in vitro assays utilizing two-dimensional monolayers of cancer cells and animal models. Additionally, animal models do not truly mimic the original human tumor, are time consuming, and usually costly. New preclinical models are needed for innovation in cancer translational research. Hence, it is time to welcome the three-dimensional (3D) organoids: self-organizing cells grown in 3D culture systems mimicking the parent tissues from which the primary cells originate. The 3D organoids offer deeper insights into the crucial cellular processes in tissue and organ formation and pathological processes. Generation of near-perfect physiological microenvironments allow 3D organoids to couple with gene editing tools, such as the clustered regularly interspersed short palindromic repeat (CRISPR)/CRISPR-associated 9 and the transcription activator-like effector nucleases to model human diseases, offering distinct advantages over current models. We explain in this expert review that through recapitulating patients' normal and tumor tissues, organoid technology can markedly advance personalized medicine and help reveal once hidden aspects of cancers. The use of defined tissue- or organ-specific matrices, among other factors, will likely allow organoid technology to realize its potential in innovating many fields of life sciences.
Collapse
Affiliation(s)
- Kevin Dzobo
- 1 International Center for Genetic Engineering and Biotechnology (ICGEB) , Cape Town Component, Cape Town, South Africa .,2 Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town , Cape Town, South Africa
| | - Arielle Rowe
- 1 International Center for Genetic Engineering and Biotechnology (ICGEB) , Cape Town Component, Cape Town, South Africa
| | - Dimakatso A Senthebane
- 1 International Center for Genetic Engineering and Biotechnology (ICGEB) , Cape Town Component, Cape Town, South Africa .,2 Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town , Cape Town, South Africa
| | - Mousa A M AlMazyadi
- 3 Al-Ahsa College of Medicine, King Faisal University , Al-Ahsa, Kingdom of Saudi Arabia
| | - Victoria Patten
- 2 Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town , Cape Town, South Africa
| | - M Iqbal Parker
- 2 Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town , Cape Town, South Africa
| |
Collapse
|
215
|
Ren H, Hou X, Eiken PW, Zhang J, Pierson KE, Nair AA, Davila JI, Kovarikova H, Jang JS, Johnson SH, Molina JR, Marks RS, Yang P, Yi JE, Mansfield AS, Jen J. Identification and Development of a Lung Adenocarcinoma PDX Model With STRN-ALK Fusion. Clin Lung Cancer 2019; 20:e142-e147. [PMID: 30581091 DOI: 10.1016/j.cllc.2018.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/29/2018] [Accepted: 11/12/2018] [Indexed: 02/05/2023]
Affiliation(s)
- Hongzheng Ren
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Biomarker Discovery Program, Mayo Clinic, Rochester, MN; Cancer Research Center, Shantou University Medical College, Shantou, China
| | - Xiaonan Hou
- Department of Medical Oncology, Mayo Clinic, Rochester, MN
| | | | - Jin Zhang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | | | - Asha A Nair
- Department of Health Science Research, Mayo Clinic, Rochester, MN
| | - Jaime I Davila
- Department of Health Science Research, Mayo Clinic, Rochester, MN
| | - Helena Kovarikova
- Institute of Clinical Biochemistry and Diagnostics, Charles University, Faculty of Medicine, Hradec Kralove, Czech Republic; Institute of Clinical Biochemistry and Diagnostics, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jin Sung Jang
- The Genome Analysis Core, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | | | | | - Ping Yang
- Department of Health Science Research, Mayo Clinic, Rochester, MN
| | - Joanne E Yi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - Jin Jen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Biomarker Discovery Program, Mayo Clinic, Rochester, MN; The Genome Analysis Core, Center for Individualized Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
216
|
Samkoe KS, Schultz E, Solanki A, Wang L, Korber J, Tichauer KM, Gibbs SL. Simultaneous extracellular and intracellular quantification of EGFR using paired-agent imaging in an in ovo tumor model. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2019; 10859. [PMID: 32296254 DOI: 10.1117/12.2510778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Quantification of protein concentrations is often a static and tissue destructive technique. Paired-agent imaging (PAI) using matched targeted and untargeted agents has been established as a dynamic method for quantifying the extracellular domain of epidermal growth factor receptor (EGFR) in vivo in a variety of tumor lines. Here we extend the PAI model to simultaneously quantify the extracellular and intracellular regions of EGFR using novel cell membrane permeable fluorescent small molecules, TRITC-erlotinib (targeted) and BODIPY-N-erlotinib (non-binding control isoform) synthesized in house. An EGFR overexpressing squamous cell carcinoma cell xenograft tumor, A431, was implanted on the chorioallantoic membrane (CAM) of the embryonated chicken egg. In total six fluorescent molecules were administered and monitored over 1 h using multi-spectral imaging. EGFR concentrations were determined using both extracellular and intracellular PAI methods. The fluorescent molecules used for extracellular PAI were ABY-029, an anti-EGFR Affibody molecule conjugated to IRDye 800CW, and a Control Imaging Agent Affibody molecule conjugated to IRDye 680RD. The intracellular PAI (iPAI) fluorescent molecules were cell membrane penetrating TRITC-erlotinib, BODIPY-N-erlotinb, and BODIPY TR carboxylate, as well as cell membrane impermeant control agent, Alexa Fluor 647 carboxylate. Results from simultaneous imaging of both the extracellular and intracellular binding domains of EGFR indicate that concentrations of intracellular EGFR are higher than extracellular. This is anticipated as EGFR exists in two distinct populations in cells, cell membrane bound and internalized, activated protein. iPAI is a promising new method for quantifying intracellular proteins in a rapid tumor model on the chicken CAM.
Collapse
Affiliation(s)
- Kimberley S Samkoe
- Geisel School of Medicine, Dartmouth College, Hanover, NH 03755.,Department of Surgery, Dartmouth-Hitchcock, Lebanon, NH, 03756
| | - Emily Schultz
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, 60616
| | - Allison Solanki
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, 60616
| | - Lei Wang
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, 60616
| | - Jesse Korber
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, 60616
| | - Kenneth M Tichauer
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97201
| | - Summer L Gibbs
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, 60616
| |
Collapse
|
217
|
Pereira NB, de Souza JC, Bastos VC, Fonseca FP, de Avelar GF, Castro WH, Dias AAM, Mosqueda‐Taylor A, Gomez RS, Gomes CC. Patient‐derived xenografts of a case of ameloblastic fibrodentinoma. Oral Dis 2019; 25:1229-1233. [DOI: 10.1111/odi.13056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/04/2019] [Accepted: 01/25/2019] [Indexed: 12/26/2022]
Affiliation(s)
- Núbia B. Pereira
- Departament of Pathology, Biological Science Institute Universidade Federal de Minas Gerais (UFMG) Belo Horizonte Brazil
| | - Juliana C. de Souza
- Departament of Pathology, Biological Science Institute Universidade Federal de Minas Gerais (UFMG) Belo Horizonte Brazil
| | - Victor C. Bastos
- Departament of Pathology, Biological Science Institute Universidade Federal de Minas Gerais (UFMG) Belo Horizonte Brazil
| | - Felipe P. Fonseca
- Departament of Oral Surgery and Pathology, School of Dentistry Universidade Federal de Minas Gerais (UFMG) Belo Horizonte Brazil
| | - Gleide F. de Avelar
- Departament of Morphology, Biological Science Institute Universidade Federal de Minas Gerais (UFMG) Belo Horizonte Brazil
| | - Wagner H. Castro
- Departament of Oral Surgery and Pathology, School of Dentistry Universidade Federal de Minas Gerais (UFMG) Belo Horizonte Brazil
| | - Adriana A. M. Dias
- Departament of General Biology, Biological Science Institute Universidade Federal de Minas Gerais (UFMG) Belo Horizonte Brazil
| | | | - Ricardo S. Gomez
- Departament of Oral Surgery and Pathology, School of Dentistry Universidade Federal de Minas Gerais (UFMG) Belo Horizonte Brazil
| | - Carolina C. Gomes
- Departament of Pathology, Biological Science Institute Universidade Federal de Minas Gerais (UFMG) Belo Horizonte Brazil
| |
Collapse
|
218
|
Mouse-Derived Isograft (MDI) In Vivo Tumor Models I. Spontaneous sMDI Models: Characterization and Cancer Therapeutic Approaches. Cancers (Basel) 2019; 11:cancers11020244. [PMID: 30791466 PMCID: PMC6406567 DOI: 10.3390/cancers11020244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 02/13/2019] [Accepted: 02/15/2019] [Indexed: 01/05/2023] Open
Abstract
Syngeneic in vivo tumor models are valuable for the development and investigation of immune-modulating anti-cancer drugs. In the present study, we established a novel syngeneic in vivo model type named mouse-derived isografts (MDIs). Spontaneous MDIs (sMDIs) were obtained during a long-term observation period (more than one to two years) of naïve and untreated animals of various mouse strains (C3H/HeJ, CBA/J, DBA/2N, BALB/c, and C57BL/6N). Primary tumors or suspicious tissues were assessed macroscopically and re-transplanted in a PDX-like manner as small tumor pieces into sex-matched syngeneic animals. Nine outgrowing primary tumors were histologically characterized either as adenocarcinomas, histiocytic carcinomas, or lymphomas. Growth of the tumor pieces after re-transplantation displayed model heterogeneity. The adenocarcinoma sMDI model JA-0009 was further characterized by flow cytometry, RNA-sequencing, and efficacy studies. M2 macrophages were found to be the main tumor infiltrating leukocyte population, whereas only a few T cells were observed. JA-0009 showed limited sensitivity when treated with antibodies against inhibitory checkpoint molecules (anti-mPD-1 and anti-mCTLA-4), but high sensitivity to gemcitabine treatment. The generated sMDI are spontaneously occurring tumors of low passage number, propagated as tissue pieces in mice without any tissue culturing, and thus conserving the original tumor characteristics and intratumoral immune cell populations.
Collapse
|
219
|
Wei X, Mao T, Li S, He J, Hou X, Li H, Zhan M, Yang X, Li R, Xiao J, Yuan S, Sun L. DT-13 inhibited the proliferation of colorectal cancer via glycolytic metabolism and AMPK/mTOR signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 54:120-131. [PMID: 30668361 DOI: 10.1016/j.phymed.2018.09.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/22/2018] [Accepted: 09/03/2018] [Indexed: 05/16/2023]
Abstract
BACKGROUND Emerging hallmark of cancer is reprogrammed cellular metabolism, increased glycolytic metabolism is physiological characteristic of human malignant neoplasms. Saponin monomer 13 of the dwarf lilyturf tuber (DT-13) is the main steroidal saponin from Liriopes Radix, which has been reported to exert anti-inflammation and anti-tumor activities but low toxicity to normal tissue. However, the effect of DT-13 on metabolism process is still unclear. PURPOSE This study aims to characterize the role of DT-13 in glucose metabolism in colorectal cancer cells, and investigate whether the metabolism process is involved in the anti-cancer response of DT-13. METHODS Colony formation assay was employed to determine anti-proliferative effect induced by DT-13 at 2.5, 5, 10 μM. Apoptosis and cell cycle arrest were detected by Annexin V/PI staining and PI staining, respectively. Genetic inhibition of glycolytic metabolism was carried out by knockdown of GLUT1. Orthotopic implantation mouse model of colorectal cancer was used to assess in vivo antitumor effect of DT-13 (0.625, 1.25, 2.5 mg/kg). The chemoprevention effect of DT-13 (10mg/kg) was evaluated by using C57BL/6J APCmin mice model. Glycolytic-related key enzymes and AMPK pathway were detected by using quantitative real-time PCR, western blotting, and immunohistochemical staining. RESULTS Our results showed that cell proliferation was significantly inhibited by DT-13 in a dose-dependent manner. DT-13 inhibited glucose uptake, ATP generation, and reduced lactate production. Furthermore, DT-13 remarkably inhibited GLUT1 expression in both mRNA and protein levels. Knocking down of GLUT1 led to reduced inhibition of glucose uptake after DT-13 treatment. Moreover, deletion of GLUT1 decreased inhibitory ratio of DT-13 on cancer growth. Orthotopic implantation mouse model of colorectal cancer further confirmed that DT-13 inhibited colorectal cancer growth via blocking GLUT1 in vivo. In addition, C57BL/6J APCmin mice model revealed that DT-13 dramatically reduced the total number of spontaneous adenomas in intestinal, which further confirmed the anti-tumor activity of DT-13 in colorectal cancer. Furthermore, the mechanistically investigation showed DT-13 activated AMPK and inhibited m-TOR to block cancer growth in vitro. CONCLUSION DT-13 is a potent anticancer agent for colorectal cancer.
Collapse
Affiliation(s)
- Xiaohui Wei
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Tingting Mao
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Sijing Li
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jinyong He
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xiaoying Hou
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Hongyang Li
- Institute of Dermatology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Meixiao Zhan
- Zhuhai Precision Medicine Center, Zhuhai People's Hospital, Zhuhai, Guangdong, China
| | - Xiangyu Yang
- Zhuhai Precision Medicine Center, Zhuhai People's Hospital, Zhuhai, Guangdong, China
| | - Ruiming Li
- Tasly Research Institute, Tianjin Tasly Holding Group Co. Ltd., Tianjin 300410, China
| | - Jing Xiao
- Zhuhai Precision Medicine Center, Zhuhai People's Hospital, Zhuhai, Guangdong, China.
| | - Shengtao Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Li Sun
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
220
|
Du H, Hirabayashi K, Ahn S, Kren NP, Montgomery SA, Wang X, Tiruthani K, Mirlekar B, Michaud D, Greene K, Herrera SG, Xu Y, Sun C, Chen Y, Ma X, Ferrone CR, Pylayeva-Gupta Y, Yeh JJ, Liu R, Savoldo B, Ferrone S, Dotti G. Antitumor Responses in the Absence of Toxicity in Solid Tumors by Targeting B7-H3 via Chimeric Antigen Receptor T Cells. Cancer Cell 2019; 35:221-237.e8. [PMID: 30753824 PMCID: PMC6645919 DOI: 10.1016/j.ccell.2019.01.002] [Citation(s) in RCA: 315] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 10/31/2018] [Accepted: 01/02/2019] [Indexed: 12/20/2022]
Abstract
The high expression across multiple tumor types and restricted expression in normal tissues make B7-H3 an attractive target for immunotherapy. We generated chimeric antigen receptor (CAR) T cells targeting B7-H3 (B7-H3.CAR-Ts) and found that B7-H3.CAR-Ts controlled the growth of pancreatic ductal adenocarcinoma, ovarian cancer and neuroblastoma in vitro and in orthotopic and metastatic xenograft mouse models, which included patient-derived xenograft. We also found that 4-1BB co-stimulation promotes lower PD-1 expression in B7-H3.CAR-Ts, and superior antitumor activity when targeting tumor cells that constitutively expressed PD-L1. We took advantage of the cross-reactivity of the B7-H3.CAR with murine B7-H3, and found that B7-H3.CAR-Ts significantly controlled tumor growth in a syngeneic tumor model without evident toxicity. These findings support the clinical development of B7-H3.CAR-Ts.
Collapse
MESH Headings
- Animals
- B7 Antigens/genetics
- B7 Antigens/immunology
- B7-H1 Antigen/immunology
- CD28 Antigens/immunology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/therapy
- Cell Line, Tumor
- Coculture Techniques
- Female
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Male
- Mice, Inbred C57BL
- Neuroblastoma/genetics
- Neuroblastoma/immunology
- Neuroblastoma/pathology
- Neuroblastoma/therapy
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Signal Transduction
- Tumor Burden
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Hongwei Du
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Koichi Hirabayashi
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sarah Ahn
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nancy Porterfield Kren
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Stephanie Ann Montgomery
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Xinhui Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Karthik Tiruthani
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Bhalchandra Mirlekar
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Daniel Michaud
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kevin Greene
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Silvia Gabriela Herrera
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yang Xu
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Chuang Sun
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yuhui Chen
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Xingcong Ma
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Cristina Rosa Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Yuliya Pylayeva-Gupta
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jen Jen Yeh
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Surgery, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rihe Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA; Carolina Center for Genome Sciences, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pediatrics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
221
|
Tu MJ, Ho PY, Zhang QY, Jian C, Qiu JX, Kim EJ, Bold RJ, Gonzalez FJ, Bi H, Yu AM. Bioengineered miRNA-1291 prodrug therapy in pancreatic cancer cells and patient-derived xenograft mouse models. Cancer Lett 2019; 442:82-90. [PMID: 30389433 PMCID: PMC6311422 DOI: 10.1016/j.canlet.2018.10.038] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/12/2018] [Accepted: 10/25/2018] [Indexed: 02/08/2023]
Abstract
Our recent studies have revealed that microRNA-1291 (miR-1291) is downregulated in pancreatic cancer (PC) specimens and restoration of miR-1291 inhibits tumorigenesis of PC cells. This study is to assess the efficacy and underlying mechanism of our bioengineered miR-1291 prodrug monotherapy and combined treatment with chemotherapy. AT-rich interacting domain protein 3B (ARID3B) was verified as a new target for miR-1291, and miR-1291 prodrug was processed to mature miR-1291 in PC cells which surprisingly upregulated ARID3B mRNA and protein levels. Co-administration of miR-1291 with gemcitabine plus nab-paclitaxel (Gem-nP) largely increased the levels of apoptosis, DNA damage and mitotic arrest in PC cells, compared to mono-drug treatment. Consequently, miR-1291 prodrug improved cell sensitivity to Gem-nP. Furthermore, systemic administration of in vivo-jetPEI-formulated miR-1291 prodrug suppressed tumor growth in both PANC-1 xenograft and PC patients derived xenograft (PDX) mouse models to comparable degrees as Gem-nP alone, while combination treatment reduced tumor growth more ubiquitously and to the greatest degrees (70-90%), compared to monotherapy. All treatments were well tolerated in mice. In conclusion, biologic miR-1291 prodrug has therapeutic potential as a monotherapy for PC, and a sensitizing agent to chemotherapy.
Collapse
Affiliation(s)
- Mei-Juan Tu
- Department of Biochemistry & Molecular Medicine, UC Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Pui Yan Ho
- Department of Biochemistry & Molecular Medicine, UC Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Qian-Yu Zhang
- Department of Biochemistry & Molecular Medicine, UC Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Chao Jian
- Department of Biochemistry & Molecular Medicine, UC Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Jing-Xin Qiu
- Roswell Park Cancer Institute, Buffalo, NY, 14263, USA
| | - Edward J Kim
- Division of Hematology and Oncology, UC Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Richard J Bold
- Department of Surgery, UC Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Huichang Bi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ai-Ming Yu
- Department of Biochemistry & Molecular Medicine, UC Davis School of Medicine, Sacramento, CA, 95817, USA.
| |
Collapse
|
222
|
Engineered bone for probing organotypic growth and therapy response of prostate cancer tumoroids in vitro. Biomaterials 2019; 197:296-304. [PMID: 30682644 DOI: 10.1016/j.biomaterials.2019.01.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 12/24/2018] [Accepted: 01/20/2019] [Indexed: 01/23/2023]
Abstract
Mechanistic analysis of metastatic prostate cancer (PCa) biology and therapy response critically depends upon clinically relevant three-dimensional (3D) bone-like, organotypic culture. We here combine an engineered bone-mimetic environment (BME) with longitudinal microscopy to test the growth and therapy response of 3D PCa tumoroids. Besides promoting both tumor-cell autonomous and microenvironment-dependent growth in PCa cell lines and patient-derived xenograft cells, the BME enables in vivo-like tumor cell response to therapy, and reveals bone stroma dependent resistance to chemotherapy and BME-targeted localization and induction of cytoxicity by Radium-223. The BME platform will allow the propagation, compound screening and mechanistic dissection of patient-derived bone tumor isolates and applications toward personalized medicine.
Collapse
|
223
|
Sato K, Niida A, Masuda T, Shimizu D, Tobo T, Kuroda Y, Eguchi H, Nakagawa T, Suzuki Y, Mimori K. Multiregion Genomic Analysis of Serially Transplanted Patient-derived Xenograft Tumors. Cancer Genomics Proteomics 2019; 16:21-27. [PMID: 30587497 PMCID: PMC6348396 DOI: 10.21873/cgp.20109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 10/31/2018] [Accepted: 11/02/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Intratumoral heterogeneity (ITH) is a major cause underlying therapeutic difficulty of cancer. Although an understanding of ITH is critically important in order to develop novel therapeutic strategies, experimental models that enable the examination of ITH in a time series are lacking. MATERIALS AND METHODS We developed an experimental approach based on patient-derived xenograft (PDX) mice and a multiregional sequencing approach (MRA). The multiple regions of primary colorectal cancer (CRC) and serially transplanted PDX tumors were analyzed via whole-exome sequencing and bioinformatic analyses. RESULTS Our PDX-MRA of CRC indicated the spatiotemporal genetic transition of ITH. It was found that the subclonal architecture of CRC dynamically changes during serial transplantation. Furthermore, our data suggest that environmental selective pressures drive the development of minor pre-existing subclones in PDX-MRA. CONCLUSION PDX-MRA is a useful tool for understanding the spatiotemporal dynamics of ITH.
Collapse
Affiliation(s)
- Kuniaki Sato
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Atsushi Niida
- Division of Health Medical Computational Science, Health Intelligence Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Takaaki Masuda
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Dai Shimizu
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Taro Tobo
- Department of Clinical Laboratory Medicine and Pathology, Kyushu University Beppu Hospital, Oita, Japan
| | - Yousuke Kuroda
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Hidetoshi Eguchi
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Takashi Nakagawa
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yutaka Suzuki
- Medical Genome Sciences, Graduate School of Frontier Sciences, University of Tokyo, Chiba, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| |
Collapse
|
224
|
Wang Z, Fu S, Zhao J, Zhao W, Shen Z, Wang D, Duan J, Bai H, Wan R, Yu J, Wang S, Chen H, Chen B, Wang L, Wang J. Transbronchoscopic patient biopsy-derived xenografts as a preclinical model to explore chemorefractory-associated pathways and biomarkers for small-cell lung cancer. Cancer Lett 2019; 440-441:180-188. [PMID: 30347283 DOI: 10.1016/j.canlet.2018.10.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/29/2018] [Accepted: 10/08/2018] [Indexed: 12/28/2022]
Abstract
Insufficient tumor tissue is a major barrier for cancer biology research in small-cell lung cancer (SCLC) and has driven the development of patient-derived xenografts (PDXs) from biopsy tumor tissues. Here, we utilized transbronchoscopic biopsy specimens from SCLC tumors to establish PDXs and evaluated the genomic profile using next-generation sequencing and an RNA sequencing platform. The PDX establishment rate was 54.1% (40/74). PDXs largely recapitulated the major characteristics of their corresponding primary tumors, such as histopathology, genetic profile, and chemo-responsiveness. Compared with chemosensitive (chemo-S) PDXs, chemorefractory (chemo-R) PDXs demonstrated significant gene aberrances in the mitogen-activated protein kinase (MAPK) pathway and a higher frequency of receptor tyrosine kinase (RTK)-related genes. Phosphorylated ERK (pERK) was associated with chemo-R status. Patients with positive pERK expression demonstrated significantly inferior progression-free survival after first-line chemotherapy compared with that of patients who were negative for pERK (p < 0.001). Collectively, transbronchoscopic biopsy SCLC PDXs can serve as a model for genomic profiling and identifying biomarkers predictive of chemo-R status. Using PDXs, RTK-related gene aberrances and pERK expression were found to be associated with chemo-R SCLC.
Collapse
Affiliation(s)
- Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuai Fu
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong University, Jinan, China
| | - Jun Zhao
- Department of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wei Zhao
- Department of Cell Biology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhirong Shen
- The BeiGene Pharmaceutical Co. Ltd., Zhongguancun Life Science Park, Beijing, China
| | - Di Wang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Jianchun Duan
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hua Bai
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Rui Wan
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiangyong Yu
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuhang Wang
- GCP Center, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hanxiao Chen
- Department of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| | - Bolu Chen
- CATS Academy Boston, 2001 Washington Street, Braintree, MA, 02184, USA
| | - Lai Wang
- The BeiGene Pharmaceutical Co. Ltd., Zhongguancun Life Science Park, Beijing, China
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
225
|
Zebrafish disease models in hematology: Highlights on biological and translational impact. Biochim Biophys Acta Mol Basis Dis 2018; 1865:620-633. [PMID: 30593895 DOI: 10.1016/j.bbadis.2018.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 02/06/2023]
Abstract
Zebrafish (Danio rerio) has proven to be a versatile and reliable in vivo experimental model to study human hematopoiesis and hematological malignancies. As vertebrates, zebrafish has significant anatomical and biological similarities to humans, including the hematopoietic system. The powerful genome editing and genome-wide forward genetic screening tools have generated models that recapitulate human malignant hematopoietic pathologies in zebrafish and unravel cellular mechanisms involved in these diseases. Moreover, the use of zebrafish models in large-scale chemical screens has allowed the identification of new molecular targets and the design of alternative therapies. In this review we summarize the recent achievements in hematological research that highlight the power of the zebrafish model for discovery of new therapeutic molecules. We believe that the model is ready to give an immediate translational impact into the clinic.
Collapse
|
226
|
Liu F, Zu X, Xie X, Liu K, Chen H, Wang T, Liu F, Bode AM, Zheng Y, Dong Z, Kim DJ. Ethyl gallate as a novel ERK1/2 inhibitor suppresses patient-derived esophageal tumor growth. Mol Carcinog 2018; 58:533-543. [PMID: 30499613 DOI: 10.1002/mc.22948] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 11/13/2018] [Accepted: 11/21/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Feifei Liu
- China-US (Henan) Hormel Cancer Institute; Henan China
| | - Xueyin Zu
- China-US (Henan) Hormel Cancer Institute; Henan China
- The Pathophysiology Department; The School of Basic Medical Sciences; Zhengzhou University; Zhengzhou Henan China
| | - Xiaomeng Xie
- China-US (Henan) Hormel Cancer Institute; Henan China
| | - Kangdong Liu
- China-US (Henan) Hormel Cancer Institute; Henan China
- The Pathophysiology Department; The School of Basic Medical Sciences; Zhengzhou University; Zhengzhou Henan China
- The Affiliated Cancer Hospital; Zhengzhou University; Zhengzhou Henan China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention; Zhengzhou Henan China
| | - Hanyong Chen
- The Hormel Institute; University of Minnesota; Austin Minnesota
| | - Ting Wang
- China-US (Henan) Hormel Cancer Institute; Henan China
| | - Fangfang Liu
- China-US (Henan) Hormel Cancer Institute; Henan China
- The Pathophysiology Department; The School of Basic Medical Sciences; Zhengzhou University; Zhengzhou Henan China
| | - Ann M. Bode
- The Hormel Institute; University of Minnesota; Austin Minnesota
| | - Yan Zheng
- The Affiliated Cancer Hospital; Zhengzhou University; Zhengzhou Henan China
| | - Zigang Dong
- China-US (Henan) Hormel Cancer Institute; Henan China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention; Zhengzhou Henan China
- The Hormel Institute; University of Minnesota; Austin Minnesota
| | - Dong Joon Kim
- China-US (Henan) Hormel Cancer Institute; Henan China
| |
Collapse
|
227
|
Fluorescent humanized anti-CEA antibody specifically labels metastatic pancreatic cancer in a patient-derived orthotopic xenograft (PDOX) mouse model. Oncotarget 2018; 9:37333-37342. [PMID: 30647873 PMCID: PMC6324662 DOI: 10.18632/oncotarget.26484] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 12/04/2018] [Indexed: 11/25/2022] Open
Abstract
Pancreatic cancer is a highly lethal disease in part due to incomplete tumor resection. Targeting by tumor-specific antibodies conjugated with a fluorescent label can result in selective labeling of cancer in vivo for surgical navigation. In the present study, we describe a patient-derived orthotopic xenograft model of pancreatic cancer that recapitulated the disease on a gross and microscopic level, along with physiologic clinical manifestations. We additionally show that the use of an anti-CEA antibody conjugated to the near-infrared (NIR) fluorescent dye, IRDye800CW, can selectively highlight the pancreatic cancer and its metastases in this model with a tumor-to-background ratio of 3.5 (SEM 0.9). The present results demonstrate the clinical potential of this labeling technique for fluorescence-guided surgery of pancreatic cancer.
Collapse
|
228
|
Chauvin A, Boisvert FM. Clinical Proteomics in Colorectal Cancer, a Promising Tool for Improving Personalised Medicine. Proteomes 2018; 6:proteomes6040049. [PMID: 30513835 PMCID: PMC6313903 DOI: 10.3390/proteomes6040049] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/22/2018] [Accepted: 11/29/2018] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer is the third most common and the fourth most lethal cancer worldwide. In most of cases, patients are diagnosed at an advanced or even metastatic stage, thus explaining the high mortality. The lack of proper clinical tests and the complicated procedures currently used for detecting this cancer, as well as for predicting the response to treatment and the outcome of a patient's resistance in guiding clinical practice, are key elements driving the search for biomarkers. In the present overview, the different biomarkers (diagnostic, prognostic, treatment resistance) discovered through proteomics studies in various colorectal cancer study models (blood, stool, biopsies), including the different proteomic techniques used for the discovery of these biomarkers, are reviewed, as well as the various tests used in clinical practice and those currently in clinical phase. These studies define the limits and perspectives related to proteomic biomarker research for personalised medicine in colorectal cancer.
Collapse
Affiliation(s)
- Anaïs Chauvin
- Department of Anatomy and Cell Biology, Université de Sherbrooke, 3201 Jean-Mignault, Sherbrooke, QC J1E 4K8, Canada.
| | - François-Michel Boisvert
- Department of Anatomy and Cell Biology, Université de Sherbrooke, 3201 Jean-Mignault, Sherbrooke, QC J1E 4K8, Canada.
| |
Collapse
|
229
|
Zhang L, Liu Z, Kong C, Liu C, Yang K, Chen H, Huang J, Qian F. Improving Drug Delivery of Micellar Paclitaxel against Non-Small Cell Lung Cancer by Coloading Itraconazole as a Micelle Stabilizer and a Tumor Vascular Manipulator. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1802112. [PMID: 30444572 DOI: 10.1002/smll.201802112] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/09/2018] [Indexed: 06/09/2023]
Abstract
Although polymeric micelles of paclitaxel (PTX) significantly reduce excipient-induced toxicity compared with Taxol, they exhibit few clinical advantages in tumor inhibition and overall survival. To improve, itraconazole (ITA), an antifungal drug with potent anti-angiogenesis activity, is co-encapsulated together with PTX within the PEG-PLA micelles. The strong intermolecular interactions between the payloads inhibit drug crystallization and prevent drugs from binding with external proteins, render super-stable micelles upon dilution and exposure to biological environment, and enter the tumor cells through endocytosis. The co-encapsulated micelles show strong anti-proliferation potency against non-small-cell lung cancer (NSCLC) and even PTX resistant NSCLC cells in vitro and significantly improve the drug accumulation within the tumor in vivo. Compared with PTX monotherapy or combination therapy using individual PTX and ITA micelles, the co-encapsulated micelle demonstrates strikingly superior efficacy in tumor growth inhibition, recurrence prevention, and reversion of PTX resistance, in Kras mutant patient derived xenografts, orthotropic models, and paclitaxel-resistance subcutaneous models. Besides the pharmacokinetic improvement, therapeutic benefits are also contributed by angiogenesis inhibition and blood vessel normalization by ITA. Utilizing the pharmaceutical and pharmacological synergies between the therapeutic agents, a simple yet effective design of a combination cancer nanomedicine that is industrially scalable and clinically translatable is achieved.
Collapse
Affiliation(s)
- Ling Zhang
- School of Pharmaceutical Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, P. R. China
| | - Zhengsheng Liu
- School of Pharmaceutical Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, P. R. China
| | - Chao Kong
- School of Pharmaceutical Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, P. R. China
| | - Chun Liu
- School of Pharmaceutical Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, P. R. China
| | - Kuan Yang
- School of Pharmaceutical Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, P. R. China
| | - Huijun Chen
- School of Pharmaceutical Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, P. R. China
| | - Jinfeng Huang
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medicine College, Beijing 100021, P. R. China
| | - Feng Qian
- School of Pharmaceutical Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, P. R. China
| |
Collapse
|
230
|
Collins AT, Lang SH. A systematic review of the validity of patient derived xenograft (PDX) models: the implications for translational research and personalised medicine. PeerJ 2018; 6:e5981. [PMID: 30498642 PMCID: PMC6252062 DOI: 10.7717/peerj.5981] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/22/2018] [Indexed: 01/11/2023] Open
Abstract
Patient-derived xenograft (PDX) models are increasingly being used in oncology drug development because they offer greater predictive value than traditional cell line models. Using novel tools to critique model validity and reliability we performed a systematic review to identify all original publications describing the derivation of PDX models of colon, prostate, breast and lung cancer. Validity was defined as the ability to recapitulate the disease of interest. The study protocol was registered with the Collaborative Approach to Meta-Analysis and Review of Animal Data from Experimental Studies (CAMARADES). Searches were performed in Embase, MEDLINE and Pubmed up to July 2017. A narrative data synthesis was performed. We identified 105 studies of model validations; 29 for breast, 29 for colon, 25 for lung, 23 for prostate and 4 for multiple tissues. 133 studies were excluded because they did not perform any validation experiments despite deriving a PDX. Only one study reported following the ARRIVE guidelines; developed to improve the standard of reporting for animal experimentation. Remarkably, half of all breast (52%) and prostate (50%) studies were judged to have high concern, in contrast to 16% of colon and 28% of lung studies. The validation criteria that most commonly failed (evidence to the contrary) were: tissue of origin not proven and histology of the xenograft not comparable to the parental tumour. Overall, most studies were categorized as unclear because one or more validation conditions were not reported, or researchers failed to provide data for a proportion of their models. For example, failure to demonstrate tissue of origin, response to standard of care agents and to exclude development of lymphoma. Validation tools have the potential to improve reproducibility, reduce waste in research and increase the success of translational studies.
Collapse
Affiliation(s)
- Anne T. Collins
- Department of Biology, University of York, York, United Kingdom
| | | |
Collapse
|
231
|
Coleman O, Henry M, O'Neill F, Roche S, Swan N, Boyle L, Murphy J, Meiller J, Conlon NT, Geoghegan J, Conlon KC, Lynch V, Straubinger NL, Straubinger RM, McVey G, Moriarty M, Meleady P, Clynes M. A Comparative Quantitative LC-MS/MS Profiling Analysis of Human Pancreatic Adenocarcinoma, Adjacent-Normal Tissue, and Patient-Derived Tumour Xenografts. Proteomes 2018; 6:proteomes6040045. [PMID: 30404163 PMCID: PMC6313850 DOI: 10.3390/proteomes6040045] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/01/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers worldwide; it develops in a relatively symptom-free manner, leading to rapid disease progression and metastasis, leading to a 5-year survival rate of less than 5%. A lack of dependable diagnostic markers and rapid development of resistance to conventional therapies are among the problems associated with management of the disease. A better understanding of pancreatic tumour biology and discovery of new potential therapeutic targets are important goals in pancreatic cancer research. This study describes the comparative quantitative LC-MS/MS proteomic analysis of the membrane-enriched proteome of 10 human pancreatic ductal adenocarcinomas, 9 matched adjacent-normal pancreas and patient-derived xenografts (PDXs) in mice (10 at F1 generation and 10 F2). Quantitative label-free LC-MS/MS data analysis identified 129 proteins upregulated, and 109 downregulated, in PDAC, compared to adjacent-normal tissue. In this study, analysing peptide MS/MS data from the xenografts, great care was taken to distinguish species-specific peptides definitively derived from human sequences, or from mice, which could not be distinguished. The human-only peptides from the PDXs are of particular value, since only human tumour cells survive, and stromal cells are replaced during engraftment in the mouse; this list is, therefore, enriched in tumour-associated proteins, some of which might be potential therapeutic or diagnostic targets. Using human-specific sequences, 32 proteins were found to be upregulated, and 113 downregulated in PDX F1 tumours, compared to primary PDAC. Differential expression of CD55 between PDAC and normal pancreas, and expression across PDX generations, was confirmed by Western blotting. These data indicate the value of using PDX models in PDAC research. This study is the first comparative proteomic analysis of PDAC which employs PDX models to identify patient tumour cell-associated proteins, in an effort to find robust targets for therapeutic treatment of PDAC.
Collapse
Affiliation(s)
- Orla Coleman
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| | - Fiona O'Neill
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| | - Sandra Roche
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| | - Niall Swan
- St. Vincent's University Hospital, Dublin 4, Ireland.
| | | | - Jean Murphy
- St. Vincent's University Hospital, Dublin 4, Ireland.
| | - Justine Meiller
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| | - Neil T Conlon
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| | | | - Kevin C Conlon
- St. Vincent's University Hospital, Dublin 4, Ireland.
- Trinity College Dublin, College Green, Dublin 2, Ireland.
| | - Vincent Lynch
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
- St. Vincent's University Hospital, Dublin 4, Ireland.
| | - Ninfa L Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA.
| | - Robert M Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA.
| | - Gerard McVey
- St. Vincent's University Hospital, Dublin 4, Ireland.
- St. Luke's Hospital, Highfield Road, Rathgar, Dublin 6, Ireland.
| | - Michael Moriarty
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
- St. Luke's Hospital, Highfield Road, Rathgar, Dublin 6, Ireland.
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| | - Martin Clynes
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| |
Collapse
|
232
|
Gahete MD, Jimenez-Vacas JM, Alors-Perez E, Herrero-Aguayo V, Fuentes-Fayos AC, Pedraza-Arevalo S, Castaño JP, Luque RM. Mouse models in endocrine tumors. J Endocrinol 2018; 240:JOE-18-0571.R1. [PMID: 30475226 DOI: 10.1530/joe-18-0571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022]
Abstract
Endocrine and neuroendocrine tumors comprise a highly heterogeneous group of neoplasms that can arise from (neuro)endocrine cells, either from endocrine glands or from the widespread diffuse neuroendocrine system, and, consequently, are widely distributed throughout the body. Due to their diversity, heterogeneity and limited incidence, studying in detail the molecular and genetic alterations that underlie their development and progression is still a highly elusive task. This, in turn, hinders the discovery of novel therapeutic options for these tumors. To circumvent these limitations, numerous mouse models of endocrine and neuroendocrine tumors have been developed, characterized and used in pre-clinical, co-clinical (implemented in mouse models and patients simultaneously) and post-clinical studies, for they represent powerful and necessary tools in basic and translational tumor biology research. Indeed, different in vivo mouse models, including cell line-based xenografts (CDXs), patient-derived xenografts (PDXs) and genetically engineered mouse models (GEMs), have been used to delineate the development, progression and behavior of human tumors. Results gained with these in vivo models have facilitated the clinical application in patients of diverse breakthrough discoveries made in this field. Herein, we review the generation, characterization and translatability of the most prominent mouse models of endocrine and neuroendocrine tumors reported to date, as well as the most relevant clinical implications obtained for each endocrine and neuroendocrine tumor type.
Collapse
Affiliation(s)
- Manuel D Gahete
- M Gahete, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, 14011, Spain
| | - Juan M Jimenez-Vacas
- J Jimenez-Vacas, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Emilia Alors-Perez
- E Alors-Perez, Department of Cell Biology, Physiology and Inmunology, Maimonides Institute for Biomedical Research of Cordoba (IMIBIC) / University of Cordoba, Cordoba, Spain
| | - Vicente Herrero-Aguayo
- V Herrero-Aguayo, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Antonio C Fuentes-Fayos
- A Fuentes-Fayos, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Sergio Pedraza-Arevalo
- S Pedraza-Arevalo, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Justo P Castaño
- J Castaño, Dpt. of Cell Biology-University of Córdoba, IMIBIC-Maimonides Biomedical Research Institute of Cordoba, Cordoba, E-14004, Spain
| | - Raul M Luque
- R Luque, Dept of Cell Biology, Phisiology and Inmunology, Section of Cell Biology, University of Cordoba, Cordoba, Spain, Cordoba, 14014, Spain
| |
Collapse
|
233
|
Lin S, Huang G, Cheng L, Li Z, Xiao Y, Deng Q, Jiang Y, Li B, Lin S, Wang S, Wu Q, Yao H, Cao S, Li Y, Liu P, Wei W, Pei D, Yao Y, Wen Z, Zhang X, Wu Y, Zhang Z, Cui S, Sun X, Qian X, Li P. Establishment of peripheral blood mononuclear cell-derived humanized lung cancer mouse models for studying efficacy of PD-L1/PD-1 targeted immunotherapy. MAbs 2018; 10:1301-1311. [PMID: 30204048 PMCID: PMC6284590 DOI: 10.1080/19420862.2018.1518948] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/23/2018] [Accepted: 08/28/2018] [Indexed: 12/15/2022] Open
Abstract
Animal models used to evaluate efficacies of immune checkpoint inhibitors are insufficient or inaccurate. We thus examined two xenograft models used for this purpose, with the aim of optimizing them. One method involves the use of peripheral blood mononuclear cells and cell line-derived xenografts (PBMCs-CDX model). For this model, we implanted human lung cancer cells into NOD-scid-IL2Rg-/- (NSI) mice, followed by injection of human PBMCs. The second method involves the use of hematopoietic stem and progenitor cells and CDX (HSPCs-CDX model). For this model, we first reconstituted the human immune system by transferring human CD34+ hematopoietic stem and progenitor cells (HSPCs-derived humanized model) and then transplanted human lung cancer cells. We found that the PBMCs-CDX model was more accurate in evaluating PD-L1/PD-1 targeted immunotherapies. In addition, it took only four weeks with the PBMCs-CDX model for efficacy evaluation, compared to 10-14 weeks with the HSPCs-CDX model. We then further established PBMCs-derived patient-derived xenografts (PDX) models, including an auto-PBMCs-PDX model using cancer and T cells from the same tumor, and applied them to assess the antitumor efficacies of anti-PD-L1 antibodies. We demonstrated that this PBMCs-derived PDX model was an invaluable tool to study the efficacies of PD-L1/PD-1 targeted cancer immunotherapies. Overall, we found our PBMCs-derived models to be excellent preclinical models for studying immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Shouheng Lin
- Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Guohua Huang
- Department of Respiratory medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lin Cheng
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhen Li
- MabSpace Biosciences Co. Ltd, Suzhou, China
| | - Yiren Xiao
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qiuhua Deng
- Department of Respiratory medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuchuan Jiang
- Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Baiheng Li
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Simiao Lin
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Suna Wang
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qiting Wu
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Huihui Yao
- Department of Outpatient, The 91th Military Hospital, Jiaozuo, China
| | - Su Cao
- Division of General Pediatrics, The 91th Military Hospital, Jiaozuo, China
| | - Yang Li
- Department of Pediatric Hematology/Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Pentao Liu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, Stem Cell and Regenerative Medicine Centre, University of Hong Kong, Hong Kong, China
| | - Wei Wei
- Guangdong Cord Blood Bank, Guangdong, China
| | - Duanqing Pei
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yao Yao
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhesheng Wen
- Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xuchao Zhang
- Guangdong Lung Cancer Institute, Medical Research Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yilong Wu
- Guangdong Lung Cancer Institute, Medical Research Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhenfeng Zhang
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuzhong Cui
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Xiaofang Sun
- Key Lab for Major Obstetric Diseases of Guangdong Province, Experimental Department of Institute of Gynaecology and Obstetrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Peng Li
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
234
|
A patient derived xenograft model of cervical cancer and cervical dysplasia. PLoS One 2018; 13:e0206539. [PMID: 30365542 PMCID: PMC6203389 DOI: 10.1371/journal.pone.0206539] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/15/2018] [Indexed: 12/27/2022] Open
Abstract
Aim To develop a patient derived xenograft (PDX) model of cervical cancer and cervical dysplasia using the subrenal capsule. Methods Cervical cancer (12 Squamous Cell Carcinoma, 1 Adenocarcinoma, 1 Adenosquamous Carcinoma), 7 cervical dysplasia biopsy and normal cervical tissues were transplanted beneath the renal capsule of immunocompromised NOD/SCID/gamma mice. Resulting tumours were harvested and portions serially transplanted into new recipient mice for up to three in vivo passages. Parent and xenograft tumours were examined by immunohistochemistry for p16INK41, HPV, and CD-45. Single cell suspensions of mixed mouse and human, or human only cell populations were also transplanted. Results The overall engraftment rate for the primary cervical cancer PDX model was 71.4 ±12.5% (n = 14). Tumours maintained morphological, histoarchitecture and immunohistochemical features of the parent tumour, and demonstrated invasiveness into local tissues. Single cell suspensions did not produce tumour growth in this model. Mean length of time (32.4 +/- 3.5 weeks) for the transplanted tissue to generate a tumour in the animal was similar between successive transplantations. Three of four xenografted cervical dysplasia tissues generated microscopic cystic structures resembling dysplastic cervical tissue. Normal cervical tissue (4 of 5 xenografted) also developed microscopic cervical tissue grafts. Conclusion The subrenal capsule can be used for a PDX model of human cervical cancer with a good engraftment rate and the ability to model in vivo characteristics of cervical cancer. For the first time we have demonstrated that cervical dysplasia and normal cervical tissue generated microscopic tissues in a PDX model.
Collapse
|
235
|
Xiong G, Stewart RL, Chen J, Gao T, Scott TL, Samayoa LM, O'Connor K, Lane AN, Xu R. Collagen prolyl 4-hydroxylase 1 is essential for HIF-1α stabilization and TNBC chemoresistance. Nat Commun 2018; 9:4456. [PMID: 30367042 PMCID: PMC6203834 DOI: 10.1038/s41467-018-06893-9] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 10/02/2018] [Indexed: 12/26/2022] Open
Abstract
Collagen prolyl 4-hydroxylase (P4H) expression and collagen hydroxylation in cancer cells are necessary for breast cancer progression. Here, we show that P4H alpha 1 subunit (P4HA1) protein expression is induced in triple-negative breast cancer (TNBC) and HER2 positive breast cancer. By modulating alpha ketoglutarate (α-KG) and succinate levels P4HA1 expression reduces proline hydroxylation on hypoxia-inducible factor (HIF) 1α, enhancing its stability in cancer cells. Activation of the P4HA/HIF-1 axis enhances cancer cell stemness, accompanied by decreased oxidative phosphorylation and reactive oxygen species (ROS) levels. Inhibition of P4HA1 sensitizes TNBC to the chemotherapeutic agent docetaxel and doxorubicin in xenografts and patient-derived models. We also show that increased P4HA1 expression correlates with short relapse-free survival in TNBC patients who received chemotherapy. These results suggest that P4HA1 promotes chemoresistance by modulating HIF-1-dependent cancer cell stemness. Targeting collagen P4H is a promising strategy to inhibit tumor progression and sensitize TNBC to chemotherapeutic agents. Hyperactivation of HIF-1α is crucial in progression of triple-negative breast cancer, but how HIF-1α stability is maintained in a hypoxia-independent manner is unclear. Here, the authors show collagen prolyl-4-hydroylase 1 stabilises HIF-1α and is involved in chemoresistance in TNBC.
Collapse
Affiliation(s)
- Gaofeng Xiong
- UK Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA.,Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA
| | - Rachel L Stewart
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Jie Chen
- UK Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA.,Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA
| | - Tianyan Gao
- UK Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA.,Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA
| | - Timothy L Scott
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, 40536, USA.,Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Luis M Samayoa
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Kathleen O'Connor
- UK Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA.,Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA
| | - Andrew N Lane
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, 40536, USA.,Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Ren Xu
- UK Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA. .,Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
236
|
Kulkarni N, Alessandrì L, Panero R, Arigoni M, Olivero M, Ferrero G, Cordero F, Beccuti M, Calogero RA. Reproducible bioinformatics project: a community for reproducible bioinformatics analysis pipelines. BMC Bioinformatics 2018; 19:349. [PMID: 30367595 PMCID: PMC6191970 DOI: 10.1186/s12859-018-2296-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Reproducibility of a research is a key element in the modern science and it is mandatory for any industrial application. It represents the ability of replicating an experiment independently by the location and the operator. Therefore, a study can be considered reproducible only if all used data are available and the exploited computational analysis workflow is clearly described. However, today for reproducing a complex bioinformatics analysis, the raw data and the list of tools used in the workflow could be not enough to guarantee the reproducibility of the results obtained. Indeed, different releases of the same tools and/or of the system libraries (exploited by such tools) might lead to sneaky reproducibility issues. Results To address this challenge, we established the Reproducible Bioinformatics Project (RBP), which is a non-profit and open-source project, whose aim is to provide a schema and an infrastructure, based on docker images and R package, to provide reproducible results in Bioinformatics. One or more Docker images are then defined for a workflow (typically one for each task), while the workflow implementation is handled via R-functions embedded in a package available at github repository. Thus, a bioinformatician participating to the project has firstly to integrate her/his workflow modules into Docker image(s) exploiting an Ubuntu docker image developed ad hoc by RPB to make easier this task. Secondly, the workflow implementation must be realized in R according to an R-skeleton function made available by RPB to guarantee homogeneity and reusability among different RPB functions. Moreover she/he has to provide the R vignette explaining the package functionality together with an example dataset which can be used to improve the user confidence in the workflow utilization. Conclusions Reproducible Bioinformatics Project provides a general schema and an infrastructure to distribute robust and reproducible workflows. Thus, it guarantees to final users the ability to repeat consistently any analysis independently by the used UNIX-like architecture.
Collapse
Affiliation(s)
- Neha Kulkarni
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Luca Alessandrì
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Riccardo Panero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Martina Olivero
- Department of Oncology, University of Torino, Candiolo, Italy
| | - Giulio Ferrero
- Department of Computer Sciences, University of Torino, Torino, Italy
| | - Francesca Cordero
- Department of Computer Sciences, University of Torino, Torino, Italy.
| | - Marco Beccuti
- Department of Computer Sciences, University of Torino, Torino, Italy
| | - Raffaele A Calogero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.
| |
Collapse
|
237
|
Drug screening of biopsy-derived spheroids using a self-generated microfluidic concentration gradient. Sci Rep 2018; 8:14672. [PMID: 30279484 PMCID: PMC6168499 DOI: 10.1038/s41598-018-33055-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 09/18/2018] [Indexed: 12/19/2022] Open
Abstract
Performing drug screening of tissue derived from cancer patient biopsies using physiologically relevant 3D tumour models presents challenges due to the limited amount of available cell material. Here, we present a microfluidic platform that enables drug screening of cancer cell-enriched multicellular spheroids derived from tumour biopsies, allowing extensive anticancer compound screening prior to treatment. This technology was validated using cell lines and then used to screen primary human prostate cancer cells, grown in 3D as a heterogeneous culture from biopsy-derived tissue. The technology enabled the formation of repeatable drug concentration gradients across an array of spheroids without external fluid actuation, delivering simultaneously a range of drug concentrations to multiple sized spheroids, as well as replicates for each concentration. As proof-of-concept screening, spheroids were generated from two patient biopsies and a panel of standard-of-care compounds for prostate cancer were tested. Brightfield and fluorescence images were analysed to provide readouts of spheroid growth and health, as well as drug efficacy over time. Overall, this technology could prove a useful tool for personalised medicine and future drug development, with the potential to provide cost- and time-reduction in the healthcare delivery.
Collapse
|
238
|
Zhao Y, Shuen TWH, Toh TB, Chan XY, Liu M, Tan SY, Fan Y, Yang H, Lyer SG, Bonney GK, Loh E, Chang KTE, Tan TC, Zhai W, Chan JKY, Chow EKH, Chee CE, Lee GH, Dan YY, Chow PKH, Toh HC, Lim SG, Chen Q. Development of a new patient-derived xenograft humanised mouse model to study human-specific tumour microenvironment and immunotherapy. Gut 2018; 67:1845-1854. [PMID: 29602780 PMCID: PMC6145285 DOI: 10.1136/gutjnl-2017-315201] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 03/07/2018] [Accepted: 03/17/2018] [Indexed: 12/27/2022]
Abstract
OBJECTIVE As the current therapeutic strategies for human hepatocellular carcinoma (HCC) have been proven to have limited effectiveness, immunotherapy becomes a compelling way to tackle the disease. We aim to provide humanised mouse (humice) models for the understanding of the interaction between human cancer and immune system, particularly for human-specific drug testing. DESIGN Patient-derived xenograft tumours are established with type I human leucocyte antigen matched human immune system in NOD-scid Il2rg-/- (NSG) mice. The longitudinal changes of the tumour and immune responses as well as the efficacy of immune checkpoint inhibitors are investigated. RESULTS Similar to the clinical outcomes, the human immune system in our model is educated by the tumour and exhibits exhaustion phenotypes such as a significant declination of leucocyte numbers, upregulation of exhaustion markers and decreased the production of human proinflammatory cytokines. Notably, cytotoxic immune cells decreased more rapidly compared with other cell types. Tumour infiltrated T cells have much higher expression of exhaustion markers and lower cytokine production compared with peripheral T cells. In addition, tumour-associated macrophages and myeloid-derived suppressor cells are found to be highly enriched in the tumour microenvironment. Interestingly, the tumour also changes gene expression profiles in response to immune responses by upregulating immune checkpoint ligands. Most importantly, in contrast to the NSG model, our model demonstrates both therapeutic and side effects of immune checkpoint inhibitors pembrolizumab and ipilimumab. CONCLUSIONS Our work provides a model for immune-oncology study and a useful parallel-to-human platform for anti-HCC drug testing, especially immunotherapy.
Collapse
Affiliation(s)
- Yue Zhao
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | | | - Tan Boon Toh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Xue Ying Chan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Min Liu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Sue Yee Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hechuan Yang
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Shridhar Ganpathi Lyer
- Division of Hepatobiliary and Liver Transplantation Surgery, National University Health System, Singapore
| | - Glenn Kunnath Bonney
- Division of Hepatobiliary and Liver Transplantation Surgery, National University Health System, Singapore
| | - Eva Loh
- Department of Pathology and Laboratory Medicine, KK Women’s and Children’s Hospital, Singapore
| | - Kenneth Tou En Chang
- Department of Pathology and Laboratory Medicine, KK Women’s and Children’s Hospital, Singapore
| | - Thiam Chye Tan
- Department of Obstetrics and Gynaecology, KK Women’s and Children’s Hospital, Singapore
| | - Weiwei Zhai
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women’s and Children’s Hospital, Singapore
- Experimental Fetal Medicine Group, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Cheng Ean Chee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Guan Huei Lee
- Division of Gastroenterology and Hepatology, National University Health System, Singapore, Singapore
| | - Yock Young Dan
- Division of Gastroenterology and Hepatology, National University Health System, Singapore, Singapore
| | - Pierce Kah-Hoe Chow
- Division of Surgical Oncology, National Cancer Center Singapore, Singapore
- Department of Hepato-Pancreato-Biliary and Transplant Surgery, Singapore General Hospital, Singapore
- Duke-NUS Graduate Medical School, Singapore
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Seng Gee Lim
- Division of Gastroenterology and Hepatology, National University Health System, Singapore, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
239
|
Two novel camptothecin derivatives inhibit colorectal cancer proliferation via induction of cell cycle arrest and apoptosis in vitro and in vivo. Eur J Pharm Sci 2018; 123:546-559. [DOI: 10.1016/j.ejps.2018.08.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/13/2018] [Accepted: 08/13/2018] [Indexed: 12/22/2022]
|
240
|
Sayles LC, Breese MR, Koehne AL, Leung SG, Lee AG, Liu HY, Spillinger A, Shah AT, Tanasa B, Straessler K, Hazard FK, Spunt SL, Marina N, Kim GE, Cho SJ, Avedian RS, Mohler DG, Kim MO, DuBois SG, Hawkins DS, Sweet-Cordero EA. Genome-Informed Targeted Therapy for Osteosarcoma. Cancer Discov 2018; 9:46-63. [PMID: 30266815 DOI: 10.1158/2159-8290.cd-17-1152] [Citation(s) in RCA: 257] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 08/01/2018] [Accepted: 09/25/2018] [Indexed: 11/16/2022]
Abstract
Osteosarcoma is a highly aggressive cancer for which treatment has remained essentially unchanged for more than 30 years. Osteosarcoma is characterized by widespread and recurrent somatic copy-number alterations (SCNA) and structural rearrangements. In contrast, few recurrent point mutations in protein-coding genes have been identified, suggesting that genes within SCNAs are key oncogenic drivers in this disease. SCNAs and structural rearrangements are highly heterogeneous across osteosarcoma cases, suggesting the need for a genome-informed approach to targeted therapy. To identify patient-specific candidate drivers, we used a simple heuristic based on degree and rank order of copy-number amplification (identified by whole-genome sequencing) and changes in gene expression as identified by RNA sequencing. Using patient-derived tumor xenografts, we demonstrate that targeting of patient-specific SCNAs leads to significant decrease in tumor burden, providing a road map for genome-informed treatment of osteosarcoma. SIGNIFICANCE: Osteosarcoma is treated with a chemotherapy regimen established 30 years ago. Although osteosarcoma is genomically complex, we hypothesized that tumor-specific dependencies could be identified within SCNAs. Using patient-derived tumor xenografts, we found a high degree of response for "genome-matched" therapies, demonstrating the utility of a targeted genome-informed approach.This article is highlighted in the In This Issue feature, p. 1.
Collapse
Affiliation(s)
- Leanne C Sayles
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Marcus R Breese
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Amanda L Koehne
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Stanley G Leung
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Alex G Lee
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Heng-Yi Liu
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Aviv Spillinger
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Avanthi T Shah
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Bogdan Tanasa
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Krystal Straessler
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Florette K Hazard
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, California
| | - Sheri L Spunt
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford University, Stanford, California
| | - Neyssa Marina
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford University, Stanford, California
| | - Grace E Kim
- Department of Pathology, University of California, San Francisco, California
| | - Soo-Jin Cho
- Department of Pathology, University of California, San Francisco, California
| | - Raffi S Avedian
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford University, Stanford, California
| | - David G Mohler
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford University, Stanford, California
| | - Mi-Ok Kim
- Biostatistics Core, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Division of Biostatistics, Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - Steven G DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center and Harvard Medical School, Boston, Massachusetts
| | - Douglas S Hawkins
- Seattle Children's Hospital, University of Washington, Fred Hutchison Cancer Research Center, Seattle, Washington
| | - E Alejandro Sweet-Cordero
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California.
| |
Collapse
|
241
|
Kim H, Schaniel C. Modeling Hematological Diseases and Cancer With Patient-Specific Induced Pluripotent Stem Cells. Front Immunol 2018; 9:2243. [PMID: 30323816 PMCID: PMC6172418 DOI: 10.3389/fimmu.2018.02243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/10/2018] [Indexed: 12/13/2022] Open
Abstract
The advent of induced pluripotent stem cells (iPSCs) together with recent advances in genome editing, microphysiological systems, tissue engineering and xenograft models present new opportunities for the investigation of hematological diseases and cancer in a patient-specific context. Here we review the progress in the field and discuss the advantages, limitations, and challenges of iPSC-based malignancy modeling. We will also discuss the use of iPSCs and its derivatives as cellular sources for drug target identification, drug development and evaluation of pharmacological responses.
Collapse
Affiliation(s)
- Huensuk Kim
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Christoph Schaniel
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
242
|
Callahan SJ, Tepan S, Zhang YM, Lindsay H, Burger A, Campbell NR, Kim IS, Hollmann TJ, Studer L, Mosimann C, White RM. Cancer modeling by Transgene Electroporation in Adult Zebrafish (TEAZ). Dis Model Mech 2018; 11:dmm.034561. [PMID: 30061297 PMCID: PMC6177007 DOI: 10.1242/dmm.034561] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/06/2018] [Indexed: 12/19/2022] Open
Abstract
Transgenic animals are invaluable for modeling cancer genomics, but often require complex crosses of multiple germline alleles to obtain the desired combinations. Zebrafish models have advantages in that transgenes can be rapidly tested by mosaic expression, but typically lack spatial and temporal control of tumor onset, which limits their utility for the study of tumor progression and metastasis. To overcome these limitations, we have developed a method referred to as Transgene Electroporation in Adult Zebrafish (TEAZ). TEAZ can deliver DNA constructs with promoter elements of interest to drive fluorophores, oncogenes or CRISPR-Cas9-based mutagenic cassettes in specific cell types. Using TEAZ, we created a highly aggressive melanoma model via Cas9-mediated inactivation of Rb1 in the context of BRAFV600E in spatially constrained melanocytes. Unlike prior models that take ∼4 months to develop, we found that TEAZ leads to tumor onset in ∼7 weeks, and these tumors develop in fully immunocompetent animals. As the resulting tumors initiated at highly defined locations, we could track their progression via fluorescence, and documented deep invasion into tissues and metastatic deposits. TEAZ can be deployed to other tissues and cell types, such as the heart, with the use of suitable transgenic promoters. The versatility of TEAZ makes it widely accessible for rapid modeling of somatic gene alterations and cancer progression at a scale not achievable in other in vivo systems.
Collapse
Affiliation(s)
- Scott J Callahan
- Memorial Sloan Kettering Cancer Center, Cancer Biology and Genetics and Department of Medicine, New York, NY 10065, USA.,Memorial Sloan Kettering Cancer Center, Developmental Biology, New York, NY 10065, USA.,Memorial Sloan Kettering Cancer Center, Gerstner Graduate School of Biomedical Sciences, New York, NY 10065, USA
| | - Stephanie Tepan
- Memorial Sloan Kettering Cancer Center, 2017 Summer Clinical Oncology Research Experience (SCORE) Program, New York, NY 10065, USA.,Hunter College, New York, NY 10065, USA
| | - Yan M Zhang
- Memorial Sloan Kettering Cancer Center, Cancer Biology and Genetics and Department of Medicine, New York, NY 10065, USA
| | - Helen Lindsay
- Institute of Molecular Life Sciences, University of Zurich, Zurich 8057, Switzerland.,SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich 8057, Switzerland
| | - Alexa Burger
- Institute of Molecular Life Sciences, University of Zurich, Zurich 8057, Switzerland
| | - Nathaniel R Campbell
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA
| | - Isabella S Kim
- Memorial Sloan Kettering Cancer Center, Cancer Biology and Genetics and Department of Medicine, New York, NY 10065, USA
| | - Travis J Hollmann
- Memorial Sloan Kettering Cancer Center, Pathology, New York, NY 10065, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan Kettering Institute, New York, NY 10065, USA; Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Christian Mosimann
- Institute of Molecular Life Sciences, University of Zurich, Zurich 8057, Switzerland
| | - Richard M White
- Memorial Sloan Kettering Cancer Center, Cancer Biology and Genetics and Department of Medicine, New York, NY 10065, USA .,Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
243
|
Chamberlain CE, German MS, Yang K, Wang J, VanBrocklin H, Regan M, Shokat KM, Ducker GS, Kim GE, Hann B, Donner DB, Warren RS, Venook AP, Bergsland EK, Lee D, Wang Y, Nakakura EK. A Patient-derived Xenograft Model of Pancreatic Neuroendocrine Tumors Identifies Sapanisertib as a Possible New Treatment for Everolimus-resistant Tumors. Mol Cancer Ther 2018; 17:2702-2709. [PMID: 30254185 DOI: 10.1158/1535-7163.mct-17-1204] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 07/18/2018] [Accepted: 09/20/2018] [Indexed: 12/11/2022]
Abstract
Patients with pancreatic neuroendocrine tumors (PNET) commonly develop advanced disease and require systemic therapy. However, treatment options remain limited, in part, because experimental models that reliably emulate PNET disease are lacking. We therefore developed a patient-derived xenograft model of PNET (PDX-PNET), which we then used to evaluate two mTOR inhibitor drugs: FDA-approved everolimus and the investigational new drug sapanisertib. PDX-PNETs maintained a PNET morphology and PNET-specific gene expression signature with serial passage. PDX-PNETs also harbored mutations in genes previously associated with PNETs (such as MEN1 and PTEN), displayed activation of the mTOR pathway, and could be detected by Gallium-68 DOTATATE PET-CT. Treatment of PDX-PNETs with either everolimus or sapanisertib strongly inhibited growth. As seen in patients, some PDX-PNETs developed resistance to everolimus. However, sapanisertib, a more potent inhibitor of the mTOR pathway, caused tumor shrinkage in most everolimus-resistant tumors. Our PDX-PNET model is the first available, validated PDX model for PNET, and preclinical data from the use of this model suggest that sapanisertib may be an effective new treatment option for patients with PNET or everolimus-resistant PNET.
Collapse
Affiliation(s)
- Chester E Chamberlain
- Center for Regeneration Medicine, University of California, San Francisco, California.
- Diabetes Center, University of California, San Francisco, California
- Department of Medicine, University of California, San Francisco, California
| | - Michael S German
- Center for Regeneration Medicine, University of California, San Francisco, California
- Diabetes Center, University of California, San Francisco, California
- Department of Medicine, University of California, San Francisco, California
| | - Katherine Yang
- Center for Regeneration Medicine, University of California, San Francisco, California
- Diabetes Center, University of California, San Francisco, California
- Department of Medicine, University of California, San Francisco, California
| | - Jason Wang
- Center for Regeneration Medicine, University of California, San Francisco, California
- Diabetes Center, University of California, San Francisco, California
- Department of Medicine, University of California, San Francisco, California
| | - Henry VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Melanie Regan
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Kevan M Shokat
- Department of Cellular Molecular Pharmacology, University of California, San Francisco, California
| | - Gregory S Ducker
- Department of Cellular Molecular Pharmacology, University of California, San Francisco, California
| | - Grace E Kim
- Department of Pathology, University of California, San Francisco, California
| | - Byron Hann
- Helen Diller Family HDF Comprehensive Cancer Center, University of California, San Francisco, California
| | - David B Donner
- Helen Diller Family HDF Comprehensive Cancer Center, University of California, San Francisco, California
- Department of Surgery, University of California, San Francisco, California
| | - Robert S Warren
- Helen Diller Family HDF Comprehensive Cancer Center, University of California, San Francisco, California
- Department of Surgery, University of California, San Francisco, California
| | - Alan P Venook
- Department of Medicine, University of California, San Francisco, California
- Helen Diller Family HDF Comprehensive Cancer Center, University of California, San Francisco, California
| | - Emily K Bergsland
- Department of Medicine, University of California, San Francisco, California
- Helen Diller Family HDF Comprehensive Cancer Center, University of California, San Francisco, California
| | - Danny Lee
- Helen Diller Family HDF Comprehensive Cancer Center, University of California, San Francisco, California
- Department of Surgery, University of California, San Francisco, California
| | - Yucheng Wang
- Helen Diller Family HDF Comprehensive Cancer Center, University of California, San Francisco, California
- Department of Surgery, University of California, San Francisco, California
| | - Eric K Nakakura
- Helen Diller Family HDF Comprehensive Cancer Center, University of California, San Francisco, California.
- Department of Surgery, University of California, San Francisco, California
| |
Collapse
|
244
|
Dang SC, Fan YY, Cui L, Chen JX, Qu JG, Gu M. PLK1 as a potential prognostic marker of gastric cancer through MEK-ERK pathway on PDTX models. Onco Targets Ther 2018; 11:6239-6247. [PMID: 30288059 PMCID: PMC6163028 DOI: 10.2147/ott.s169880] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background PLK1 has been identified as having a great effect on cell division and maintaining genomic stability in mitosis, spindle assembly, and DNA damage response by current studies. Materials and methods We assessed PLK1 expression in cervical cancer tissues and cells. We have also evaluated the effects of PLK1 on gastric cancer cell proliferation, migration, and apoptosis both in vitro and in vivo. Results Our results show that PLK1 is overexpressed in gastric cancer tissues and cells. Inhibition of PLK1 contributes cell cycle G2-phase arrest and inhibits the proliferation, migration, and apoptosis of gastric cancer (GC) cells, whereas its overexpression promotes proliferation, migration, and apoptosis in these cells. Moreover, PLK1 inhibition reduces expression of pMEK and pERK. More importantly, in vivo by analyzing tumorigenesis in patient-derived tumor xenograft (PDTX) models, the inhibition of PLK1 activity by BI6727 significantly decreased the volume and weight of the tumors compared with control group (P<0.01). Conclusion Our results found that PLK1 has a significant impact on the survival of GC cells; it may become a prognostic judge, a potential therapeutic target, and a preventative biomarker of GC.
Collapse
Affiliation(s)
- Sheng-Chun Dang
- Department of General Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Yi-Yi Fan
- Department of General Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Lei Cui
- Department of General Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Ji-Xiang Chen
- Department of General Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Jian-Guo Qu
- Department of General Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Min Gu
- Zhenjiang Integrative Medicine Hospital, Zhenjiang, Jiangsu Province, People's Republic of China,
| |
Collapse
|
245
|
Wilson JJ, Chow KH, Labrie NJ, Branca JA, Sproule TJ, Perkins BRA, Wolf EE, Costa M, Stafford G, Rosales C, Mills KD, Roopenian DC, Hasham MG. Enhancing the efficacy of glycolytic blockade in cancer cells via RAD51 inhibition. Cancer Biol Ther 2018; 20:169-182. [PMID: 30183475 PMCID: PMC6343731 DOI: 10.1080/15384047.2018.1507666] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Targeting the early steps of the glycolysis pathway in cancers is a well-established therapeutic strategy; however, the doses required to elicit a therapeutic effect on the cancer can be toxic to the patient. Consequently, numerous preclinical and clinical studies have combined glycolytic blockade with other therapies. However, most of these other therapies do not specifically target cancer cells, and thus adversely affect normal tissue. Here we first show that a diverse number of cancer models – spontaneous, patient-derived xenografted tumor samples, and xenografted human cancer cells – can be efficiently targeted by 2-deoxy-D-Glucose (2DG), a well-known glycolytic inhibitor. Next, we tested the cancer-cell specificity of a therapeutic compound using the MEC1 cell line, a chronic lymphocytic leukemia (CLL) cell line that expresses activation induced cytidine deaminase (AID). We show that MEC1 cells, are susceptible to 4,4ʹ-Diisothiocyano-2,2ʹ-stilbenedisulfonic acid (DIDS), a specific RAD51 inhibitor. We then combine 2DG and DIDS, each at a lower dose and demonstrate that this combination is more efficacious than fludarabine, the current standard- of- care treatment for CLL. This suggests that the therapeutic blockade of glycolysis together with the therapeutic inhibition of RAD51-dependent homologous recombination can be a potentially beneficial combination for targeting AID positive cancer cells with minimal adverse effects on normal tissue. Implications: Combination therapy targeting glycolysis and specific RAD51 function shows increased efficacy as compared to standard of care treatments in leukemias.
Collapse
Affiliation(s)
- John J Wilson
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Kin-Hoe Chow
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Nathan J Labrie
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Jane A Branca
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Thomas J Sproule
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Bryant R A Perkins
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Elise E Wolf
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Mauro Costa
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Grace Stafford
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Christine Rosales
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | | | - Derry C Roopenian
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Muneer G Hasham
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| |
Collapse
|
246
|
Patient-Derived Xenograft Models for Endometrial Cancer Research. Int J Mol Sci 2018; 19:ijms19082431. [PMID: 30126113 PMCID: PMC6121639 DOI: 10.3390/ijms19082431] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/03/2018] [Accepted: 08/13/2018] [Indexed: 12/21/2022] Open
Abstract
Endometrial cancer (EC) is the most common malignancy of the genital tract among women in developed countries. Recently, a molecular classification of EC has been performed providing a system that, in conjunction with histological observations, reliably improves EC classification and enhances patient management. Patient-derived xenograft models (PDX) represent nowadays a promising tool for translational research, since they closely resemble patient tumour features and retain molecular and histological features. In EC, PDX models have already been used, mainly as an individualized approach to evaluate the efficacy of novel therapies and to identify treatment-response biomarkers; however, their uses in more global or holistic approaches are still missing. As a collaborative effort within the ENITEC network, here we describe one of the most extensive EC PDX cohorts developed from primary tumour and metastasis covering all EC subtypes. Our models are histologically and molecularly characterized and represent an excellent reservoir of EC tumour samples for translational research. This review compiles the information on current methods of EC PDX generation and their utility and provides new perspectives for the exploitation of these valuable tools in order to increase the success ratio for translating results to clinical practice.
Collapse
|
247
|
Guo S, Gao S, Liu R, Shen J, Shi X, Bai S, Wang H, Zheng K, Shao Z, Liang C, Peng S, Jin G. Oncological and genetic factors impacting PDX model construction with NSG mice in pancreatic cancer. FASEB J 2018; 33:873-884. [PMID: 30091943 DOI: 10.1096/fj.201800617r] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A patient-derived xenograft (PDX) approach, which relies on direct transplantation of tumor specimens into an immunocompromised animal, is a commonly used method for investigating tumor therapy predictions in vivo. This study evaluated influencing factors, including clinical, oncological, and genetic variables, for a pancreatic PDX model in mice. Tumor specimens were obtained from 121 patients with pancreatic ductal adenocarcinoma who underwent surgical resection at the Changhai Pancreatic Surgery Medical Center (Shanghai, China) between April 2016 and February 2017. Pancreatic cancer (PC) samples <3 mm3 were subcutaneously implanted into the NOD/Shi-scid/IL-2Rγnull (NSG) mice. Once the xenograft reached 300-500 mm3 or reached 180 d after cell inoculation, the tumor was excised. Part of the tumor was subsequently transplanted to next-generation mice, and another part was analyzed by using immunohistochemistry. Among the 121 patients with PC, tumor xenograft was successfully generated in 86 patients (71.1%). Primary tumor >3.5 cm in size was independently associated with xenograft formation rate. In addition, several enriched mutated genes within the VEGF pathway and higher microvessel density were found in the positive group (with xenograft) compared with the negative group (without xenograft). We concluded that tumor size and mutated VEGF pathway in PC are important factors affecting PDX model construction with NSG mice.-Guo, S., Gao, S., Liu, R., Shen, J., Shi, X., Bai, S., Wang, H., Zheng, K., Shao, Z., Liang, C., Peng, S., Jin, G. Oncological and genetic factors impacting PDX model construction with NSG mice in pancreatic cancer.
Collapse
Affiliation(s)
- Shiwei Guo
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China; and
| | - Suizhi Gao
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China; and
| | - Rendong Liu
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China; and
| | - Jing Shen
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China; and
| | - Xiaohan Shi
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China; and
| | - Sijia Bai
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China; and
| | - Huan Wang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China; and
| | - Kailian Zheng
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China; and
| | - Zhuo Shao
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China; and
| | | | - Siying Peng
- Beijing IDMO Company Limited, Beijing, China
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China; and
| |
Collapse
|
248
|
Van Nyen T, Moiola CP, Colas E, Annibali D, Amant F. Modeling Endometrial Cancer: Past, Present, and Future. Int J Mol Sci 2018; 19:E2348. [PMID: 30096949 PMCID: PMC6121384 DOI: 10.3390/ijms19082348] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 12/13/2022] Open
Abstract
Endometrial cancer is the most common type of cancer of the female reproductive tract. Although prognosis is generally good for patients with low-grade and early-stage diseases, the outcomes for high-grade and metastatic/recurrent cases remain poor, since traditional chemotherapy regimens based on platinum and taxanes have limited effects. No targeted agents have been approved so far, although several new drugs have been tested without striking results in clinical trials. Over the last decades, many efforts have been made towards the establishment and development of preclinical models, aiming at recapitulating the structural and molecular determinants of the disease. Here, we present an overview of the most commonly used in vitro and in vivo models and discuss their peculiar features, describing their main applications and the value in the advancement of both fundamental and translational endometrial cancer research.
Collapse
Affiliation(s)
- Tom Van Nyen
- Department of Oncology, Gynecological Oncology, KU Leuven, 3000 Leuven, Belgium.
| | - Cristian P Moiola
- Pathological Oncology Group, Biomedical Research Institute of Lleida (IRBLLEIDA), University Hospital Arnau de Vilanova, 25198 Lleida, Spain.
- Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, CIBERONC, 08035 Barcelona, Spain.
| | - Eva Colas
- Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, CIBERONC, 08035 Barcelona, Spain.
| | - Daniela Annibali
- Department of Oncology, Gynecological Oncology, KU Leuven, 3000 Leuven, Belgium.
| | - Frédéric Amant
- Department of Oncology, Gynecological Oncology, KU Leuven, 3000 Leuven, Belgium.
- Centre for Gynecologic Oncology Amsterdam (CGOA), Antoni Van Leeuwenhoek-Netherlands Cancer Institute (Avl-NKI) and University Medical Centra (UMC), 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
249
|
Dai W, Liu J, Li Q, Liu W, Li YX, Li YY. A comparison of next-generation sequencing analysis methods for cancer xenograft samples. J Genet Genomics 2018; 45:345-350. [PMID: 30055875 DOI: 10.1016/j.jgg.2018.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/15/2018] [Accepted: 07/09/2018] [Indexed: 12/13/2022]
Abstract
The application of next-generation sequencing (NGS) technology in cancer is influenced by the quality and purity of tissue samples. This issue is especially critical for patient-derived xenograft (PDX) models, which have proven to be by far the best preclinical tool for investigating human tumor biology, because the sensitivity and specificity of NGS analysis in xenograft samples would be compromised by the contamination of mouse DNA and RNA. This definitely affects downstream analyses by causing inaccurate mutation calling and gene expression estimates. The reliability of NGS data analysis for cancer xenograft samples is therefore highly dependent on whether the sequencing reads derived from the xenograft could be distinguished from those originated from the host. That is, each sequence read needs to be accurately assigned to its original species. Here, we review currently available methodologies in this field, including Xenome, Disambiguate, bamcmp and pdxBlacklist, and provide guidelines for users.
Collapse
Affiliation(s)
- Wentao Dai
- Shanghai Center for Bioinformation Technology, Shanghai 201203, China; Shanghai Engineering Research Center of Pharmaceutical Translation & Shanghai Industrial Technology Institute, Shanghai 201203, China; Shanghai Industrial Technology Institute, Shanghai 201203, China
| | - Jixiang Liu
- Shanghai Center for Bioinformation Technology, Shanghai 201203, China; Shanghai Engineering Research Center of Pharmaceutical Translation & Shanghai Industrial Technology Institute, Shanghai 201203, China; Shanghai Industrial Technology Institute, Shanghai 201203, China
| | - Quanxue Li
- Shanghai Center for Bioinformation Technology, Shanghai 201203, China; School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Wei Liu
- Shanghai Center for Bioinformation Technology, Shanghai 201203, China; Shanghai Engineering Research Center of Pharmaceutical Translation & Shanghai Industrial Technology Institute, Shanghai 201203, China; Shanghai Industrial Technology Institute, Shanghai 201203, China
| | - Yi-Xue Li
- Shanghai Center for Bioinformation Technology, Shanghai 201203, China; Shanghai Engineering Research Center of Pharmaceutical Translation & Shanghai Industrial Technology Institute, Shanghai 201203, China; School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; Shanghai Industrial Technology Institute, Shanghai 201203, China.
| | - Yuan-Yuan Li
- Shanghai Center for Bioinformation Technology, Shanghai 201203, China; Shanghai Engineering Research Center of Pharmaceutical Translation & Shanghai Industrial Technology Institute, Shanghai 201203, China; Shanghai Industrial Technology Institute, Shanghai 201203, China.
| |
Collapse
|
250
|
Patient-derived xenograft cryopreservation and reanimation outcomes are dependent on cryoprotectant type. J Transl Med 2018; 98:947-956. [PMID: 29520054 PMCID: PMC6072591 DOI: 10.1038/s41374-018-0042-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 01/14/2018] [Accepted: 02/16/2018] [Indexed: 01/22/2023] Open
Abstract
Patient-derived xenografts (PDX) are being increasingly utilized in preclinical oncologic research. Maintaining large colonies of early generation tumor-bearing mice is impractical and cost-prohibitive. Optimal methods for efficient long-term cryopreservation and subsequent reanimation of PDX tumors are critical to any viable PDX program. We sought to compare the performance of "Standard" and "Specialized" cryoprotectant media on various cryopreservation and reanimation outcomes in PDX tumors. Standard (10% DMSO media) and Specialized (Cryostor®) media were compared between overall and matched PDX tumors. Primary outcome was reanimation engraftment efficiency (REE). Secondary outcomes included time to tumor formation (TTF), time to harvest (TTH), and potential loss of unique PDX lines. Overall 57 unique PDX tumors underwent 484 reanimation engraftment attempts after previous cryopreservation. There were 10 unique PDX tumors cryopreserved with Standard (71 attempts), 40 with Specialized (272 attempts), and 7 with both (141 attempts). Median frozen time of reanimated tumors was 29 weeks (max. 177). Tumor pathology, original primary PDX growth rates, frozen storage times, and number of implantations per PDX model were similar between cryoprotectant groups. Specialized media resulted in superior REE (overall: 82 vs. 39%, p < 0.0001; matched: 97 vs. 36%, p < 0.0001; >52 weeks cryostorage: 59 vs. 9%, p < 0.0001), shorter TTF (overall 24 vs. 54 days, p = 0.0051; matched 18 vs. 53 days, p = 0.0013) and shorter TTH (overall: 64 vs. 89 days, p = 0.009; matched: 47 vs. 88 days, p = 0.0005) compared to Standard. Specialized media demonstrated improved REE with extended duration cryostorage (p = 0.048) compared to Standard. Potential loss of unique PDX lines was lower with Specialized media (9 vs. 35%, p = 0.017). In conclusion, cryopreservation with a specialized cryoprotectant appears superior to traditional laboratory-based media and can be performed with reliable reanimation even after extended cryostorage.
Collapse
|