1
|
Pu F, Guo H, Shi D, Chen F, Peng Y, Huang X, Liu J, Zhang Z, Shao Z. The generation and use of animal models of osteosarcoma in cancer research. Genes Dis 2024; 11:664-674. [PMID: 37692517 PMCID: PMC10491873 DOI: 10.1016/j.gendis.2022.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 12/16/2022] [Indexed: 09/12/2023] Open
Abstract
Osteosarcoma is the most common malignant bone tumor affecting children and adolescents. Currently, the most common treatment is surgery combined with neoadjuvant chemotherapy. Although the survival rate of patients with osteosarcoma has improved in recent years, it remains poor when the tumor(s) progress and distant metastases develop. Therefore, better animal models that more accurately replicate the natural progression of the disease are needed to develop improved prognostic and diagnostic markers, as well as targeted therapies for both primary and metastatic osteosarcoma. The present review described animal models currently being used in research investigating osteosarcoma, and their characteristics, advantages, and disadvantages. These models may help elucidate the pathogenic mechanism(s) of osteosarcoma and provide evidence to support and develop clinical treatment strategies.
Collapse
Affiliation(s)
- Feifei Pu
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine (Wuhan No.1 Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Haoyu Guo
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Deyao Shi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Fengxia Chen
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, China
- Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, China
| | - Yizhong Peng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xin Huang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jianxiang Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Zhicai Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| |
Collapse
|
2
|
Isaacson N, Lopez-Ambrosio K, Chubb L, Waanders N, Hoffmann E, Witt C, James S, Prawel DA. Compressive properties and failure behavior of photocast hydroxyapatite gyroid scaffolds vary with porosity. J Biomater Appl 2022; 37:55-76. [PMID: 35331033 DOI: 10.1177/08853282211073904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Hydroxyapatite is commonly used in tissue engineered scaffolds for bone regeneration due to its excellent bioactivity and slow degradation rate in the human body. A method of layer-wise, photopolymerized viscous extrusion, a type of additive manufacturing, was developed to fabricate hydroxyapatite gyroid scaffolds with 60%, 70%, and 80% porosities. This study uses this method to produce and evaluate calcium phosphate-based scaffolds. Gyroid topology was selected due to its interconnected porosity and superior, isotropic mechanical properties compared to typical rectilinear lattice structures. These 3D printed scaffolds were mechanically tested in compression and examined to determine the relationship between porosity, ultimate compressive strength, and fracture behavior. Compressive strength increased with decreasing porosity. Ultimate compressive strengths of the 60% and 70% porous gyroids are comparable to that of human cancellous bone, and higher than previously reported for hydroxyapatite rectilinear scaffolds. These gyroid scaffolds exhibited ultimate compressive strength increases between 1.5 and 6.5 times greater than expected, based on volume of material, as porosity is decreased. The Weibull moduli, a measure of failure predictability, were predictive of failure mode and found to be in the accepted range for engineering ceramics. The gyroid scaffolds were also found to be self-reinforcing such that initial failures due to minor manufacturing inconsistencies did not appear to be the primary cause of early failure of the scaffold. The porous gyroids exhibited scaffold failure characteristics that varied with porosity, ranging from monolithic failure to layer-by-layer failure, and demonstrated self-reinforcement in each porosity tested.
Collapse
Affiliation(s)
- Nelson Isaacson
- School of Advanced Materials Discovery, 3447Colorado State University, Fort Collins, CO, USA
| | | | - Laura Chubb
- College of Veterinary Medicine and Biomedical Sciences, 3447Colorado State University, Fort Collins, CO, USA
| | - Nathan Waanders
- School of Biomedical Engineering, 3447Colorado State University, Fort Collins, CO, USA
| | - Emily Hoffmann
- School of Biomedical Engineering, 3447Colorado State University, Fort Collins, CO, USA
| | - Connor Witt
- Department of Chemical and Biological Engineering, 3447Colorado State University, Fort Collins, CO, USA
| | - Susan James
- School of Advanced Materials Discovery, 3447Colorado State University, Fort Collins, CO, USA.,School of Biomedical Engineering, 3447Colorado State University, Fort Collins, CO, USA.,Mechanical Engineering, 3447Colorado State University, Fort Collins, CO, USA
| | - David A Prawel
- School of Advanced Materials Discovery, 3447Colorado State University, Fort Collins, CO, USA.,School of Biomedical Engineering, 3447Colorado State University, Fort Collins, CO, USA.,Mechanical Engineering, 3447Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
3
|
Wu H, He Z, Li X, Xu X, Zhong W, Bu J, Huang G. Efficient and Consistent Orthotopic Osteosarcoma Model by Cell Sheet Transplantation in the Nude Mice for Drug Testing. Front Bioeng Biotechnol 2021; 9:690409. [PMID: 34631675 PMCID: PMC8498338 DOI: 10.3389/fbioe.2021.690409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma is a big challenge on clinical treatment. The breakthrough associated with osteosarcoma in basic research and translational research depends on the reliable establishment of an animal model, whereby mice are frequently used. However, a traditional animal modeling technique like tumor cell suspension injection causes batch dynamics and large mice consumption. Here, we suggested a novel approach in establishing an orthotropic osteosarcoma model in nude mice rapidly by cell sheet culture and transplantation. Our findings demonstrated that the 143b osteosarcoma cell sheet orthotopically implanted into the nude mice could form a visible mass within 10 days, whereas it took over 15 days for a similar amount of cell suspension injection to form a visible tumor mass. Living animal imaging results showed that a tumor formation rate was 100% in the cell sheet implantation group, while it was 67% in the cell suspension injection group. The formed tumor masses were highly consistent in both growth rate and tumor size. Massive bone destruction and soft tissue mass formation were observed from the micro CT analysis, suggesting the presence of osteosarcoma. The histopathological analysis demonstrated that the orthotropic osteosarcoma model mimicked the tumor bone growth, bone destruction, and the lung metastasis. These findings imply that such a cell sheet technology could be an appropriate approach to rapidly establish a sustainable orthotropic osteosarcoma model for tumor research and reduce mice consumption.
Collapse
Affiliation(s)
- Hongwei Wu
- Department of Orthopedics, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhengxi He
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, China
| | - Xianan Li
- Department of Orthopedics, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xuezheng Xu
- Department of Orthopedics, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wu Zhong
- Department of Orthopedics, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jie Bu
- Department of Orthopedics, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Gang Huang
- Department of Orthopedics, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
4
|
Tumor cell MT1-MMP is dispensable for osteosarcoma tumor growth, bone degradation and lung metastasis. Sci Rep 2020; 10:19138. [PMID: 33154487 PMCID: PMC7645741 DOI: 10.1038/s41598-020-75995-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/16/2020] [Indexed: 02/03/2023] Open
Abstract
The membrane-anchored matrix metalloprotease MT1-MMP is a potent collagenolytic enzyme with a well-established role in extracellular matrix turnover and cellular invasion into collagen-rich tissues. MT1-MMP is highly expressed in various types of cancer and has been demonstrated to be directly involved in several stages of tumor progression, including primary tumor growth, angiogenesis, invasion and metastasis. Osteosarcoma is the most common type of primary bone cancer. This disease is characterized by invasive tumor growth, leading to extensive bone destruction, and metastasis to the lungs. The tumor cells in human osteosarcoma display a strong expression of MT1-MMP, but the role of MT1-MMP in osteosarcoma progression is currently unknown. In this study, we investigated the role of MT1-MMP during various stages of osteosarcoma development. We utilized an optimized orthotopic murine osteosarcoma model and human osteosarcoma cells in which the MT1-MMP gene was knocked out using CRISPR/Cas9. We observed a strong expression of MT1-MMP in wildtype cells of both primary tumors and lung metastases, but, surprisingly, MT1-MMP deficiency did not affect primary tumor growth, bone degradation or the formation and growth of lung metastases. We therefore propose that, unlike findings reported in other cancers, tumor-expressed MT1-MMP is dispensable for all stages of osteosarcoma progression.
Collapse
|
5
|
McCann TS, Parrish JK, Hsieh J, Sechler M, Sobral LM, Self C, Jones KL, Goodspeed A, Costello JC, Jedlicka P. KDM5A and PHF2 positively control expression of pro-metastatic genes repressed by EWS/Fli1, and promote growth and metastatic properties in Ewing sarcoma. Oncotarget 2020; 11:3818-3831. [PMID: 33196691 PMCID: PMC7597412 DOI: 10.18632/oncotarget.27737] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023] Open
Abstract
Ewing sarcoma is an aggressive malignant neoplasm with high propensity for metastasis and poor clinical outcomes. The EWS/Fli1 oncofusion is the disease driver in > 90% of cases, but presents a difficult therapeutic target. Moreover, EWS/Fli1 plays a complex role in disease progression, with inhibitory effects on critical steps of metastasis. Like many other pediatric cancers, Ewing sarcoma is a disease marked by epigenetic dysregulation. Epigenetic mechanisms present alternative targeting opportunities, but their contributions to Ewing sarcoma metastasis and disease progression remain poorly understood. Here, we show that the epigenetic regulators KDM5A and PHF2 promote growth and metastatic properties in Ewing sarcoma, and, strikingly, activate expression many pro-metastatic genes repressed by EWS/Fli1. These genes include L1CAM, which is associated with adverse outcomes in Ewing sarcoma, and promotes migratory and invasive properties. KDM5A and PHF2 retain their growth promoting effects in more metastatically potent EWS/Fli1low cells, and PHF2 promotes both invasion and L1CAM expression in this cell population. Furthermore, KDM5A and PHF2 each contribute to the increased metastatic potency of EWS/Fli1low cells in vivo. Together, these studies identify KDM5A and PHF2 as novel disease-promoting factors, and potential new targets, in Ewing sarcoma, including the more metastatically potent EWS/Fli1low cell population.
Collapse
Affiliation(s)
- Tyler S McCann
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Janet K Parrish
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Joseph Hsieh
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Medical Scientist Training Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Cancer Biology Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Marybeth Sechler
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Cancer Biology Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Lays M Sobral
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Chelsea Self
- Department of Pediatrics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Kenneth L Jones
- Department of Pediatrics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Bioinformatics Shared Resource, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew Goodspeed
- Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Bioinformatics Shared Resource, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA
| | - James C Costello
- Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Bioinformatics Shared Resource, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA
| | - Paul Jedlicka
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Medical Scientist Training Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Cancer Biology Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
6
|
Maloney C, Edelman MC, Kallis MP, Soffer SZ, Symons M, Steinberg BM. Intratibial Injection Causes Direct Pulmonary Seeding of Osteosarcoma Cells and Is Not a Spontaneous Model of Metastasis: A Mouse Osteosarcoma Model. Clin Orthop Relat Res 2018; 476:1514-1522. [PMID: 29601385 PMCID: PMC6437576 DOI: 10.1007/s11999.0000000000000291] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/08/2018] [Indexed: 01/31/2023]
Abstract
BACKGROUND Although metastasis is the major cause of mortality in patients with osteosarcoma, little is known about how micrometastases progress to gross metastatic disease. Clinically relevant animal models are necessary to facilitate development of new therapies to target indolent pulmonary metastases. Intratibial injection of human and murine osteosarcoma cell lines have been described as orthotopic models that develop spontaneous pulmonary metastasis over time. However, there is variability in reported injection techniques and metastatic efficiency. QUESTIONS/PURPOSES We aimed to characterize a widely used murine model of metastatic osteosarcoma, determine whether it is appropriate to study spontaneous pulmonary metastasis by establishing a reliable volume for intratibial injection, determine the incidence of primary tumor and metastatic formation, determine the kinetics of pulmonary metastatic seeding and outgrowth, and the contribution of the primary tumor to subsequent development of metastasis. METHODS The metastatic mouse osteosarcoma cell line K7M2 was injected into the tibia of mice. The maximum volume that could be injected without leakage was determined using Evan's blue dye (n = 8 mice). Primary tumor formation and metastatic efficiency were determined by measuring the incidence of primary tumor and metastatic formation 4 weeks after intratibial injection (n = 30). The kinetics of metastatic development were determined by performing serial euthanasia at 1, 2, 3, and 4 weeks after injection (n = 24; five to six mice per group). Number of metastatic foci/histologic lung section and metastatic burden/lung section (average surface area of metastatic lesions divided by the total surface area of the lung) was calculated in a blinded fashion. To test the contribution of the primary tumor to subsequent metastases, amputations were performed 30 minutes, 4 hours, or 24 hours after injection (n = 21; five to six mice per group). Mice were euthanized after 4 weeks and metastatic burden calculated as described previously, comparing mice that had undergone amputation with control, nonamputated mice. Differences between groups were calculated using Kruskal-Wallis and one-way analysis of variance. RESULTS The maximum volume of cell suspension that could be injected without leakage was 10 μL. Intratibial injection of tumor cells led to intramedullary tumor formation in 93% of mice by 4 weeks and resulted in detectable pulmonary metastases in 100% of these mice as early as 1 week post-injection. Metastatic burden increased over time (0.88% ± 0.58, week 1; 6.6% ± 5.3, week 2; 16.1% ± 12.5, week 3; and 40.3% ± 14.83, week 4) with a mean difference from week 1 to week 4 of -39.38 (p < 0.001; 95% confidence interval [CI], -57.39 to -21.37), showing pulmonary metastatic growth over time. In contrast, the mean number of metastatic foci did not increase from week 1 to week 4 (36.4 ± 33.6 versus 49.3 ± 26.3, p = 0.18). Amputation of the injected limb at 30 minutes, 4 hours, and 24 hours after injection did not affect pulmonary metastatic burden at 4 weeks, with amputation as early as 30 minutes post-injection resulting in a metastatic burden equivalent to tumor-bearing controls (48.9% ± 6.1% versus 40.9% ± 15.3%, mean difference 7.96, p = 0.819; 95% CI, -33.9 to 18.0). CONCLUSIONS There is immediate seeding of the metastatic site after intratibial injection of the K7M2 osteosarcoma cell line, independent of a primary tumor. This is therefore not a model of spontaneous metastasis. CLINICAL RELEVANCE This model should not be used to study the early components of the metastatic cascade, but rather used as an experimental model of metastasis. Improved understanding of this commonly used model will allow for proper interpretation of existing data and inform the design of future studies exploring the biology of metastasis in osteosarcoma.
Collapse
Affiliation(s)
- Caroline Maloney
- C. Maloney, M. P. Kallis, M. Symons, B. M. Steinberg, The Elmezzi Graduate School of Molecular Medicine, Northwell Health, Manhasset, NY, USA C. Maloney, M. P. Kallis, S. Z. Soffer, M. Symons, B. M. Steinberg, Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA C. Maloney, M. P. Kallis, S. Z. Soffer, Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA M. C. Edelman, Department of Pathology and Laboratory Medicine, Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, NY, USA
| | | | | | | | | | | |
Collapse
|
7
|
Grisez BT, Ray JJ, Bostian PA, Markel JE, Lindsey BA. Highly metastatic K7M2 cell line: A novel murine model capable of in vivo imaging via luciferase vector transfection. J Orthop Res 2018; 36:10.1002/jor.23868. [PMID: 29427436 PMCID: PMC6086764 DOI: 10.1002/jor.23868] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 01/24/2018] [Indexed: 02/04/2023]
Abstract
Osteosarcoma is rare and little improvement in survival rates has occurred in the last 25 years despite modern chemotherapeutic treatment. Bioluminescent cell lines for the modeling of osteosarcoma have shown success in tracking metastases in vivo, but commonly use adenoviral vectors to transfect the native cell line with bioluminescent reporters. The purpose of this study was to develop an orthotopic model for metastatic osteosarcoma capable of in vivo monitoring of metastatic and primary tumor burden in an immunocompetent mouse and compare that model to its wild type pathogenesis. K7M2 cells were transfected using a plasmid vector and were stable after 12 weeks. Thirty-four female BALB/c mice aged 4-5 weeks underwent orthotopic implantation of either wild type (n = 12) or transfected (n = 22) K7M2 cells in the proximal tibia. Mice were monitored for tumor growth and weekly In Vivo Imaging System (IVIS) imaging was performed to monitor for pulmonary metastasis. Although tumors developed sooner in the wild type group, no significant differences were seen compared to Transfected Group 1 in rate of inoculation, growth rates after first detection, metastatic rate, and time between inoculation and death. This study establishes a new murine model for metastatic osteosarcoma using the K7M2-wt cell line transfected with a non-viral plasmid luciferase vector. The benefits of this preclinical model include an intact immune system and orthotopically driven metastatic disease; this model appears comparable to its wild type counterpart. In the future, the model may be used to examine promising immunomodulatory therapies using bioluminescence in vivo. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Brian T Grisez
- Department of Orthopaedics, West Virginia University, PO Box 9196, Morgantown, West Virginia 26506-9196
| | - Justin J Ray
- Department of Orthopaedics, West Virginia University, PO Box 9196, Morgantown, West Virginia 26506-9196
| | - Phillip A Bostian
- Department of Orthopaedics, West Virginia University, PO Box 9196, Morgantown, West Virginia 26506-9196
| | - Justin E Markel
- Department of Orthopaedics, West Virginia University, PO Box 9196, Morgantown, West Virginia 26506-9196
| | - Brock A Lindsey
- Department of Orthopaedics, West Virginia University, PO Box 9196, Morgantown, West Virginia 26506-9196
| |
Collapse
|
8
|
Aanstoos ME, Regan DP, Rose RJ, Chubb LS, Ehrhart NP. Do Mesenchymal Stromal Cells Influence Microscopic Residual or Metastatic Osteosarcoma in a Murine Model? Clin Orthop Relat Res 2016; 474:707-15. [PMID: 26018200 PMCID: PMC4746149 DOI: 10.1007/s11999-015-4362-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) have been shown in rodent models to promote primary and pulmonary metastatic sarcoma growth when injected in the presence of gross tumor. In theory, this would limit their use in a clinical setting after limb salvage treatment for osteosarcoma. Although concerning, these models do not translate to the clinical setting wherein MSCs could be used after primary tumor resection to aid in bone healing and incorporation of tumor endoprostheses. If we can determine whether the use of MSCs in this setting is safe, it might improve our ability to augment bone healing in patients undergoing limb salvage. QUESTIONS/PURPOSES The purpose of this study was to determine (1) whether MSCs promote pulmonary metastatic disease progression in a murine osteosarcoma model; and/or (2) whether they affect local disease recurrence in the presence of microscopic residual osteosarcoma. METHODS An orthotopic model of luciferase-expressing osteosarcoma was developed. At 10 days, resection of the primary tumor was performed. One hundred fourteen female C3H mice were inoculated with DLM8-luc osteosarcoma in the proximal tibia. Ninety-four mice developed orthotopic osteosarcoma with luciferase expression. Mice with bioluminescent evidence of a primary tumor received either a microscopically "clean" amputation at a time when residual microscopic metastatic disease was present in the lungs (pulmonary metastasis group; n = 65) or a "dirty" amputation (local recurrence group; n = 29). Mice were randomized to receive intravenous MSCs, MSCs at the surgical site, or no MSCs. Mice were monitored for development and progression of pulmonary metastasis and local recurrence by bioluminescence imaging and daily measurements at the surgical site. The number of pulmonary nodules, time to first evidence of metastasis, and size of recurrent tumor were compared using Kruskal-Wallis, analysis of variance, Welch's, t-tests, or Mann-Whitney tests as appropriate for the specific data sets with p < 0.05 considered significant. RESULTS Mice receiving intravenous MSCs had a faster time to first detection of pulmonary metastasis (2.93 ± 1.90 days) compared with mice with local injection of MSCs (6.94 ± 6.78 days) or no MSCs (5.93 ± 4.55 days) (p = 0.022). MSC treatment did not influence whether mice developed local recurrence (p = 0.749) or size of recurrent tumors (p = 0.221). CONCLUSIONS MSCs delivered to the surgical site did not promote local recurrence or size of recurrent tumors, but intravenous injection of MSCs did hasten onset of detection of pulmonary metastatic disease. Although local administration of MSCs into a surgical site does not appear to promote either pulmonary metastatic disease or local recurrence, large variation within groups and small numbers diminished statistical power such that a Type II error cannot be ruled out. CLINICAL RELEVANCE If MSCs are to be used to augment bone healing in the postlimb salvage setting in patients with osteosarcoma, it will be important to understand their influence, if any, on pulmonary micrometastsis or residual microscopic local disease. Although murine models do not completely recapitulate the clinical scenario, these results suggest that intravenous delivery of MSCs may promote micrometastatic pulmonary disease. Local administration into a surgical wound, even in the presence of residual microscopic disease, may be safe, at least in this murine model, but further investigation is warranted before considering the use of MSCs for clinical use in patients with osteosarcoma.
Collapse
Affiliation(s)
- Megan E. Aanstoos
- />School of Biomedical Engineering, Colorado State University, Fort Collins, CO USA
| | - Daniel P. Regan
- />Department of Microbiology, Immunology, & Pathology, Colorado State University, Fort Collins, CO USA
| | - Ruth J. Rose
- />Department of Clinical Sciences, Colorado State University, Fort Collins, CO USA
| | - Laura S. Chubb
- />Department of Clinical Sciences, Colorado State University, Fort Collins, CO USA
| | - Nicole P. Ehrhart
- />Department of Clinical Sciences and School of Biomedical Engineering, Colorado State University, 300 W Drake Road, Fort Collins, CO 80525 USA
| |
Collapse
|
9
|
Sottnik JL, Campbell B, Mehra R, Behbahani-Nejad O, Hall CL, Keller ET. Osteocytes serve as a progenitor cell of osteosarcoma. J Cell Biochem 2015; 115:1420-9. [PMID: 24700678 DOI: 10.1002/jcb.24793] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 02/19/2014] [Indexed: 01/20/2023]
Abstract
Osteosarcoma (OSA) is the most common primary bone tumor in humans. However, the cell of origin of OSA is not clearly defined although there is evidence that osteoblasts may serve as OSA progenitors. The role of osteocytes, terminally differentiated osteoblasts, as OSA progenitors has yet to be described. Analysis of patient cDNA from publicly available microarray data revealed that patients with OSA have increased expression of dentin matrix phosphoprotein 1 (DMP1), a marker of osteocytes. Analysis of multiple murine, human, and canine OSA cell lines revealed DMP1 expression. To test the tumorigenic potential of osteocytes, MLO-Y4, a SV-40 immortalized murine osteocyte cell line, was injected into subcutaneous and orthotopic (intratibial) sites of mice. Tumor growth occurred in both locations. Orthotopic MLO-Y4 tumors produced mixed osteoblastic/osteolytic radiographic lesions; a hallmark of OSA. Together, these data demonstrate for the first time that osteocytes can serve as OSA progenitors.
Collapse
Affiliation(s)
- Joseph L Sottnik
- Department of Urology, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | | | | | | | | | | |
Collapse
|
10
|
Fenger JM, London CA, Kisseberth WC. Canine osteosarcoma: a naturally occurring disease to inform pediatric oncology. ILAR J 2015; 55:69-85. [PMID: 24936031 DOI: 10.1093/ilar/ilu009] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Osteosarcoma (OSA) is the most common form of malignant bone cancer in children and dogs, although the disease occurs in dogs approximately 10 times more frequently than in people. Multidrug chemotherapy and aggressive surgical techniques have improved survival; however, new therapies for OSA are critical, as little improvement in survival times has been achieved in either dogs or people over the past 15 years, even with significant efforts directed at the incorporation of novel therapeutic approaches. Both clinical and molecular evidence suggests that human and canine OSA share many key features, including tumor location, presence of microscopic metastatic disease at diagnosis, development of chemotherapy-resistant metastases, and altered expression/activation of several proteins (e.g. Met, ezrin, phosphatase and tensin homolog, signal transducer and activator of transcription 3), and p53 mutations, among others. Additionally, canine and pediatric OSA exhibit overlapping transcriptional profiles and shared DNA copy number aberrations, supporting the notion that these diseases are similar at the molecular level. This review will discuss the similarities between pediatric and canine OSA with regard to histology, biologic behavior, and molecular genetic alterations that indicate canine OSA is a relevant, spontaneous, large animal model of the pediatric disease and outline how the study of naturally occurring OSA in dogs will offer additional insights into the biology and future treatment of this disease in both children and dogs.
Collapse
|
11
|
Fahim DK, Tatsui CE, Suki D, Gumin J, Lang FF, Rhines LD. Orthotopic murine model of a primary malignant bone tumor in the spine: functional, bioluminescence, and histological correlations. J Neurosurg Spine 2014; 21:378-85. [PMID: 24971476 DOI: 10.3171/2014.5.spine13205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECT There is currently no reproducible animal model of human primary malignant bone tumors in the spine to permit laboratory investigation of the human disease. Therefore, the authors sought to adapt their previously developed orthotopic model of spinal metastasis to a model for primary malignant bone tumors of the spine. METHODS A transperitoneal surgical approach was used to implant osteosarcoma (Krib-1) into the L-3 vertebral body of nude mice via a drill hole. Motor function was evaluated daily using the previously validated qualitative key milestones of tail dragging, dorsal stepping, hindlimb sweeping, and paralysis. A subset of these animals was euthanized upon reaching the various milestones, and the spines were removed, sectioned, and stained. The degree of spinal cord compression was correlated with the occurrence of milestones and assessed by a ratio between the neural elements divided by the area of the spinal canal. Another subset of animals received stably transfected Krib-1 cells with the luciferase gene, and bioluminescence was measured at 10, 20, and 30 days postimplantation. RESULTS Osteosarcoma xenografts grew in all animals according to a reliable and reproducible time course; the mean time for development of behavioral milestones was noted in relation to the day of implantation (Day 1). Tail dragging (Milestone 1) occurred on Day 19.06 (95% CI 16.11-22.01), dorsal stepping (Milestone 2) occurred on Day 28.78 (95% CI 26.79-30.77), hindlimb sweeping (Milestone 3) occurred on Day 35.61 (95% CI 32.9-38.32), and paralysis of the hindlimb (Milestone 4) occurred on Day 41.78 (95% CI 39.31-44.25). These clinically observed milestones correlated with increasing compression of the spinal cord on histological sections. The authors observed a progressive increase in the local bioluminescence (in photons/cm²/sec) of the implanted level over time with a mean of 2.17 (range 0.0-8.61) at Day 10, mean 4.68 (range 1.17-8.52) at Day 20, and mean 5.54 (range 1.22-9.99) at Day 30. CONCLUSIONS The authors have developed the first orthotopic murine model of a primary malignant bone tumor in the spine, in which neurological decline reproducibly correlates with tumor progression as evidenced by pathological confirmation and noninvasive bioluminescence measurements. Although developed for osteosarcoma, this model can be expanded to study other types of primary malignant bone tumors in the spine. This model will potentially allow animal testing of targeted therapies against specific primary malignant tumor types.
Collapse
Affiliation(s)
- Daniel K Fahim
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | | |
Collapse
|
12
|
Sampson VB, Kamara DF, Kolb EA. Xenograft and genetically engineered mouse model systems of osteosarcoma and Ewing's sarcoma: tumor models for cancer drug discovery. Expert Opin Drug Discov 2013; 8:1181-9. [PMID: 23844615 DOI: 10.1517/17460441.2013.817988] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION There are > 75 histological types of solid tumors that are classified into two major groups: bone and soft-tissue sarcomas. These diseases are more prevalent in children, and pediatric sarcomas tend to be highly aggressive and rapidly progressive. Sarcomas in adults may follow a more indolent course, but aggressive tumors are also common. Sarcomas that are metastatic at diagnosis, or recurrent following therapy, remain refractory to current treatment options with dismal overall survival rates. A major focus of clinical trials, for patients with sarcoma, is to identify novel and more effective therapeutic strategies targeted to genomic or proteomic aberrations specific to the malignant cells. Critical to the understanding of the potential for targeted therapies are models of disease that are representative of clinical disease and predictive of relevant clinical responses. AREAS COVERED In this article, the authors discuss the use of mouse xenograft models and genetically engineered mice in cancer drug discovery. The authors provide a special focus on models for the two most common bone sarcomas: osteosarcoma (OS) and Ewing's sarcoma (ES). EXPERT OPINION Predicting whether a new anticancer agent will have a positive therapeutic index in patients with OS and ES remains a challenge. The use of mouse sarcoma models for understanding the mechanisms involved in the response of tumors to new treatments is an important step in the process of drug discovery and the development of clinically relevant therapeutic strategies for these diseases.
Collapse
Affiliation(s)
- Valerie B Sampson
- A.I. duPont Hospital for Children, Cancer Therapeutics Laboratory , 1701 Rockland Rd, Wilmington DE, 19803 , USA
| | | | | |
Collapse
|
13
|
Garimella R, Eskew J, Bhamidi P, Vielhauer G, Hong Y, Anderson HC, Tawfik O, Rowe P. Biological characterization of preclinical Bioluminescent Osteosarcoma Orthotopic Mouse (BOOM) model: A multi-modality approach. J Bone Oncol 2013; 2:11-21. [PMID: 25688332 PMCID: PMC4327846 DOI: 10.1016/j.jbo.2012.12.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Osteosarcoma (OS) is a bone malignancy that affects children and adolescents. It is a highly aggressive tumor and typically metastasizes to lungs. Despite aggressive chemotherapy and surgical treatments, the current 5 year survival rate is 60–70%. Clinically relevant models are needed to understand OS pathobiology, metastatic progression from bones to lungs, and ultimately, to develop more efficacious treatment strategies and improve survival rates in OS patients with metastasis. The main goal of this study was to develop and characterize an in vivo OS model that will allow non-invasive tracking of tumor progression in real time, and aid in studying OS pathobiology, and screening of potential therapeutic agents against OS. In this study, we have used a multi-modality approach using bioluminescent imaging, electron microscopy, micro-computed tomography, and histopathology to develop and characterize a preclinical Bioluminescent Osteosarcoma Orthotopic Mouse (BOOM) model, using 143B human OS cell line. The results of this study clearly demonstrate that the BOOM model represents the clinical disease as evidenced by a spectrum of changes associated with tumor establishment, progression and metastasis, and detection of known OS biomarkers in the primary and metastatic tumor tissue. Key novel findings of this study include: (a) multimodality approach for extensive characterization of the BOOM model using 143B human OS cell line; (b) evidence of renal metastasis in OS orthotopic model using 143B cells; (c) evidence of Runx2 expression in the metastatic lung tissue; and (d) evidence of the presence of extracellular membrane vesicles and myofibroblasts in the BOOM model.
Collapse
Affiliation(s)
- Rama Garimella
- Division of Hematology and Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA ; Division of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS, USA ; Division of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jeff Eskew
- The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Priyanka Bhamidi
- Division of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - George Vielhauer
- The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA ; Division of Urology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Yan Hong
- Division of Gynecology and Obstetrics, University of Kansas Medical Center, Kansas City, KS, USA
| | - H Clarke Anderson
- Division of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ossama Tawfik
- Division of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Peter Rowe
- Division of Nephrology, Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
14
|
Inubushi M, Jin YN, Murai C, Hata H, Kitagawa Y, Saga T. Single-photon emission computed tomography of spontaneous liver metastasis from orthotopically implanted human colon cancer cell line stably expressing human sodium/iodide symporter reporter gene. EJNMMI Res 2012; 2:46. [PMID: 22953701 PMCID: PMC3560220 DOI: 10.1186/2191-219x-2-46] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 08/23/2012] [Indexed: 11/10/2022] Open
Abstract
UNLABELLED BACKGROUND We aimed to develop a mouse spontaneous liver metastasis model from an orthotopically implanted human colon cancer cell line stably expressing a human sodium/iodide symporter (NIS) reporter gene, which can be imaged with single-photon emission computed tomography (SPECT) using 99mTcO4-. METHODS A recombinant plasmid containing a constitutively driven NIS gene (pcDNA3-NIS) was transfected into the human colon cancer cell line HCT116, and stable cell lines were established. The stable cells were subcutaneously injected into the nude mice. When the diameter reached 10 mm, the xenografts were excised, cut into small fragments, and orthotopically implanted into the cecal walls of another nude mice. 99mTcO4- SPECT/CT imaging was initiated 8 weeks later and repeated every 1 to 2 weeks. RESULTS The production and function of NIS protein was confirmed in vitro by Western blotting and 99mTcO4- uptake assay. On SPECT/CT imaging, focal 99mTcO4- uptake was detected in the liver. Necropsy revealed local growth of the orthotopic colon xenografts with extensive invasion, microscopic serosal metastasis, and metastatic foci in the corresponding hepatic regions showing focal 99mTcO4- uptake. Immunohistochemistry revealed high levels of NIS expression in cells forming liver tumor, indicating that the liver tumor cells originated from the orthotopic colon xenografts. CONCLUSIONS The present proof-of-concept study provided a rationale for employing a radionuclide reporter gene for the specific visualization of spontaneous liver metastasis in living mice. This unique animal model of clinically relevant and externally detectable liver metastasis will be a powerful tool for investigating tumor biology and developing novel therapies for cancer metastasis.
Collapse
Affiliation(s)
- Masayuki Inubushi
- Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.
| | | | | | | | | | | |
Collapse
|
15
|
Glycolysis inhibition by 2-deoxy-d-glucose reverts the metastatic phenotype in vitro and in vivo. Clin Exp Metastasis 2011; 28:865-75. [DOI: 10.1007/s10585-011-9417-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 07/29/2011] [Indexed: 10/17/2022]
|
16
|
Abstract
Appendicular osteosarcoma (OS) is a primary mesenchymal tumor arising from malignantly transformed osteoblasts. In people, OS is the most common nonhematopoietic, primary skeletal neoplasm diagnosed in adolescents and is the second leading cause of cancer-related fatalities within this age group. Despite aggressive therapeutic management, including limb-sparing surgeries and dose-intense systemic chemotherapies, 30-40% of patients will experience progressive metastatic disease within 5 years of diagnosis. In order to reduce the fatality rate associated with recurrent or metastatic OS, a more thorough understanding of OS pathogenesis and biology is required. Towards this pursuit, comparative animal models of OS have been developed and are actively being studied to expand our fundamental understanding of OS. It is anticipated that specific animal models of OS, which most accurately recapitulate the natural disease process in people, will be most useful for advancing our understanding of OS biology, and will facilitate the discovery of disease pathogenesis and the identification of novel therapeutic strategies for managing this lethal metastatic bone sarcoma.
Collapse
Affiliation(s)
- Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, 1008 West Hazelwood Drive, Urbana, IL 61802, USA.
| |
Collapse
|
17
|
Day CP, Carter J, Bonomi C, Hollingshead M, Merlino G. Preclinical therapeutic response of residual metastatic disease is distinct from its primary tumor of origin. Int J Cancer 2011; 130:190-9. [PMID: 21312195 DOI: 10.1002/ijc.25978] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 01/05/2011] [Indexed: 12/13/2022]
Abstract
Cancer-related deaths are caused principally by recurrence and metastasis arising from residual disease, whose therapeutic responses has been suggested to be substantially different from primary tumors. However, experimental animal models designed for evaluating the therapeutic responses of residual disease are mostly lacking. To overcome this deficiency, we have developed a preclinical model that recapitulates the progression for advanced nonsmall cell lung cancer (NSCLC). An archived Lewis lung carcinoma mouse tumor, propagated only through serial in vivo transplantation and never adapted to cell culture, was stably labeled using lentivirus-encoded biomarkers, consistently expressed through an RNA polymerase II promoter. Labeled tumors were inoculated into syngeneic immunocompetent mice to ensure superior tumor-host interactions. Primary tumors were resected on reaching a predetermined size, followed by treatment in a setting akin to postsurgical first-line adjuvant chemotherapy and routine imaging to monitor the progression of pulmonary metastasis. We discovered that efficacious treatment, instead of reducing disease growth rates, significantly prolonged disease-free survival and overall survival. As in the clinic, cisplatin-based regimes were more effective in this model. However, the response of metastases to specific agents could not be predicted from, and often opposed, their effects on subcutaneous "primary" tumors, possibly due to their distinct growth kinetics and host interactions. We here introduce a clinically relevant model of residual metastatic disease that may more accurately predict the therapeutic response of recurrent, metastatic disease.
Collapse
Affiliation(s)
- Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264, USA
| | | | | | | | | |
Collapse
|
18
|
Noh K, Kim KO, Patel NR, Staples JR, Minematsu H, Nair K, Young-In Lee F. Targeting inflammatory kinase as an adjuvant treatment for osteosarcomas. J Bone Joint Surg Am 2011; 93:723-32. [PMID: 21508279 PMCID: PMC6882535 DOI: 10.2106/jbjs.j.00302] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND A subset of patients with aggressive osteosarcomas responds poorly to conventional cytotoxic chemotherapy. Recent evidence from studies involving the liver, skin, stomach, and colon suggests that carcinogenesis is associated with inflammation. Mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase 1/2 (ERK1/2) has diverse roles in cancer and inflammation. The hypothesis of the present study is that targeted ERK1/2 inhibition will demonstrate anti-cancer effects in osteosarcoma both in vitro and in vivo. METHODS The therapeutic effect of PD98059, a MAPK/ERK pathway inhibitor, was examined with respect to cell death, survival, and anti-apoptotic protein expression by means of flow cytometry and immunoblotting in vitro. Additionally, we transplanted green fluorescent protein and luciferase-tagged 143B osteosarcoma cells into the proximal part of the tibia of nude mice. Mice were randomly assigned to treatment with doxorubicin, PD98059, or both. Vehicle-treated mice served as controls. Treatment outcome was assessed by measuring bioluminescence and by monitoring survival. RESULTS In vitro, ERK1/2 blockage increased the expression of pro-apoptotic proteins and increased cell death in 143B osteosarcoma cells. Doxorubicin treatment increased the expression of Bcl-2, an anti-apoptotic protein, but this upregulation was blocked by combined treatment with PD98059, suggesting a role for ERK1/2 in conferring drug resistance. In osteosarcoma-bearing mice, targeting ERK1/2 with PD98059 resulted in prolonged survival in comparison with vehicle-treated control mice (median survival time, sixty-seven days compared with seventy-four days; p = 0.0272; survival ratio = 0.9122; 95% confidence interval = 0.4354 to 1.389). Standalone doxorubicin treatment yielded similar animal morbidity (median survival time, sixty-seven days compared with seventy-six days; p = 0.0170; survival ratio = 0.8882; 95% confidence interval = 0.4181 to 1.358). Combined PD98059 and doxorubicin treatment further prolonged survival (median survival time, sixty-seven days compared with eighty-two days; p = 0.0023; survival ratio = 0.8232; 95% confidence interval = 0.3606 to 1.286). CONCLUSIONS Inhibiting ERK1/2 signaling resulted in osteosarcoma cell death by upregulating pro-apoptotic genes and inhibiting the Bcl-2-mediated resistance to doxorubicin. In osteosarcoma-bearing mice, ERK1/2 targeting alone or in combination with doxorubicin prolonged survival as compared with untreated mice.
Collapse
Affiliation(s)
- Kyucheol Noh
- Center for Orthopaedic Research, Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street, New York, NY 10032. E-mail address for F.Y.-I. Lee:
| | - Kyung-Ok Kim
- Center for Orthopaedic Research, Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street, New York, NY 10032. E-mail address for F.Y.-I. Lee:
| | - Neel R. Patel
- Center for Orthopaedic Research, Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street, New York, NY 10032. E-mail address for F.Y.-I. Lee:
| | - J. Robert Staples
- Center for Orthopaedic Research, Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street, New York, NY 10032. E-mail address for F.Y.-I. Lee:
| | - Hiroshi Minematsu
- Center for Orthopaedic Research, Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street, New York, NY 10032. E-mail address for F.Y.-I. Lee:
| | - Kumar Nair
- Center for Orthopaedic Research, Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street, New York, NY 10032. E-mail address for F.Y.-I. Lee:
| | - Francis Young-In Lee
- Center for Orthopaedic Research, Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street, New York, NY 10032. E-mail address for F.Y.-I. Lee:
| |
Collapse
|
19
|
Wittenburg LA, Gustafson DL. Optimizing preclinical study design in oncology research. Chem Biol Interact 2011; 190:73-8. [PMID: 21296059 DOI: 10.1016/j.cbi.2011.01.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 01/07/2011] [Accepted: 01/26/2011] [Indexed: 10/18/2022]
Abstract
The current drug development pathway in oncology research has led to a large attrition rate for new drugs, in part due to a general lack of appropriate preclinical studies that are capable of accurately predicting efficacy and/or toxicity in the target population. Because of an obvious need for novel therapeutics in many types of cancer, new compounds are being investigated in human Phase I and Phase II clinical trials before a complete understanding of their toxicity and efficacy profiles is obtained. In fact, for newer targeted molecular agents that are often cytostatic in nature, the conventional preclinical evaluation used for traditional cytotoxic chemotherapies utilizing primary tumor shrinkage as an endpoint may not be appropriate. By utilizing an integrated pharmacokinetic/pharmacodynamic approach, along with proper selection of a model system, the drug development process in oncology research may be improved leading to a better understanding of the determinants of efficacy and toxicity, and ultimately fewer drugs that fail once they reach human clinical trials.
Collapse
Affiliation(s)
- Luke A Wittenburg
- Flint Animal Cancer Center, Department of Clinical Sciences, Colorado State University, 300 West Drake Road, Fort Collins, CO 80523-1620, United States.
| | | |
Collapse
|