1
|
Charbonneau M, Harper K, Brochu-Gaudreau K, Perreault A, McDonald PP, Ekindi-Ndongo N, Jeldres C, Dubois CM. Establishment of a ccRCC patient-derived chick chorioallantoic membrane model for drug testing. Front Med (Lausanne) 2022; 9:1003914. [PMID: 36275794 PMCID: PMC9582329 DOI: 10.3389/fmed.2022.1003914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is an aggressive subtype of renal cell carcinoma accounting for the majority of deaths in kidney cancer patients. Advanced ccRCC has a high mortality rate as most patients progress and develop resistance to currently approved targeted therapies, highlighting the ongoing need for adequate drug testing models to develop novel therapies. Current animal models are expensive and time-consuming. In this study, we investigated the use of the chick chorioallantoic membrane (CAM), a rapid and cost-effective model, as a complementary drug testing model for ccRCC. Our results indicated that tumor samples from ccRCC patients can be successfully cultivated on the chick chorioallantoic membrane (CAM) within 7 days while retaining their histopathological characteristics. Furthermore, treatment of ccRCC xenografts with sunitinib, a tyrosine kinase inhibitor used for the treatment of metastatic RCC, allowed us to evaluate differential responses of individual patients. Our results indicate that the CAM model is a complementary in vivo model that allows for rapid and cost-effective evaluation of ccRCC patient response to drug therapy. Therefore, this model has the potential to become a useful platform for preclinical evaluation of new targeted therapies for the treatment of ccRCC.
Collapse
Affiliation(s)
- Martine Charbonneau
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Kelly Harper
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Karine Brochu-Gaudreau
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Alexis Perreault
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | - Claudio Jeldres
- Division of Urology, Department of Surgery, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Claire M. Dubois
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada,*Correspondence: Claire M. Dubois
| |
Collapse
|
2
|
Abdolahi S, Ghazvinian Z, Muhammadnejad S, Saleh M, Asadzadeh Aghdaei H, Baghaei K. Patient-derived xenograft (PDX) models, applications and challenges in cancer research. J Transl Med 2022; 20:206. [PMID: 35538576 PMCID: PMC9088152 DOI: 10.1186/s12967-022-03405-8] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/24/2022] [Indexed: 12/12/2022] Open
Abstract
The establishing of the first cancer models created a new perspective on the identification and evaluation of new anti-cancer therapies in preclinical studies. Patient-derived xenograft models are created by tumor tissue engraftment. These models accurately represent the biology and heterogeneity of different cancers and recapitulate tumor microenvironment. These features have made it a reliable model along with the development of humanized models. Therefore, they are used in many studies, such as the development of anti-cancer drugs, co-clinical trials, personalized medicine, immunotherapy, and PDX biobanks. This review summarizes patient-derived xenograft models development procedures, drug development applications in various cancers, challenges and limitations.
Collapse
Affiliation(s)
- Shahrokh Abdolahi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeinab Ghazvinian
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Samad Muhammadnejad
- Cell-Based Therapies Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahshid Saleh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Shapiro DD, Virumbrales-Muñoz M, Beebe DJ, Abel EJ. Models of Renal Cell Carcinoma Used to Investigate Molecular Mechanisms and Develop New Therapeutics. Front Oncol 2022; 12:871252. [PMID: 35463327 PMCID: PMC9022005 DOI: 10.3389/fonc.2022.871252] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/10/2022] [Indexed: 12/24/2022] Open
Abstract
Modeling renal cell carcinoma is critical to investigating tumor biology and therapeutic mechanisms. Multiple systems have been developed to represent critical components of the tumor and its surrounding microenvironment. Prominent in vitro models include traditional cell cultures, 3D organoid models, and microphysiological devices. In vivo models consist of murine patient derived xenografts or genetically engineered mice. Each system has unique advantages as well as limitations and researchers must thoroughly understand each model to properly investigate research questions. This review addresses common model systems for renal cell carcinoma and critically evaluates their performance and ability to measure tumor characteristics.
Collapse
Affiliation(s)
- Daniel D Shapiro
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States.,Division of Urology, William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
| | - Maria Virumbrales-Muñoz
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States.,Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - David J Beebe
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States.,Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, WI, United States
| | - E Jason Abel
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
4
|
Beserra AO, Estevan EC, Bezerra SM, Torrezan GT, Ikegami A, Dellê H, Cunha IW, Meira IT, Carraro DM, Lara PN, Zequi SC, Martins VR, Santos TG. Patient-Derived Renal Cell Carcinoma Xenografts Capture Tumor Genetic Profiles and Aggressive Behaviors. KIDNEY CANCER 2022. [DOI: 10.3233/kca-210011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Patient-derived xenografts (PDX) have emerged as one of the most promising model systems to study cancer biology and to develop new antineoplastic drugs. Renal cell carcinoma (RCC) represents up to 90% of all kidney tumors, exhibits aggressive behavior, and has a propensity for metastasis. At diagnosis, 30% of patients with RCC have metastases, while up to 50% of those with localized disease treated with curative protocols experience recurrence. OBJECTIVE: This study aimed to establish an RCC PDX platform to identify novel clinical and molecular biomarkers of recurrence risk in order to facilitate precision medicine. METHODS: Tumor samples were obtained from surgical specimens of 87 RCC patients; fragments were implanted in immunodeficient NOD/SCID/gamma (NSG) mice. Seventeen fragments were implanted subcutaneously in an initial group while a second group of 70 samples were implanted orthotopically in the subcapsular space. RESULTS: A total of 19 PDX developed only after orthotopic implantation, and included 15 cases of clear cell RCC subtype, 3 cases of papillary subtype, and one unclassifiable tumor. One PDX of clear cell RCC recapitulated the phenotype of vena caval tumor thrombus extension that had been diagnosed in the source patient. PDX characterization by immunohistochemistry and targeted sequencing indicated that all PDXs preserved RCC identity and major molecular alterations. Moreover, the capacity of tumor engraftment was a strong prognostic indicator for patients with locally advanced disease. CONCLUSION: Taken together, these results suggest that the orthotopic xenograft model of RCC represents a suitable tool to study RCC biology, identify biomarkers, and to test therapeutic candidates.
Collapse
Affiliation(s)
- Adriano O. Beserra
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| | - Ethiene C. Estevan
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| | | | - Giovana T. Torrezan
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| | - Amanda Ikegami
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| | - Humberto Dellê
- Graduate Program in Medicine, Universidade Nove de Julho, São Paulo – Brazil
| | - Isabela W. Cunha
- Institute of Pathology, Rede D’OR-São Luiz and D’Or Institute for Research and Education (IDOR), São Paulo – Brazil
| | - Isabella T. Meira
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| | - Dirce M. Carraro
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| | - Primo N. Lara
- University of California Davis Comprehensive Cancer Center, Sacramento, CA – USA
| | - Stenio C. Zequi
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
- Reference Center of Urology, A.C. Camargo Cancer Center, São Paulo – Brazil
- LARCG -Latin American Renal Cancer Group
| | - Vilma R. Martins
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| | - Tiago G. Santos
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| |
Collapse
|
5
|
Esparza-López J, Longoria O, De La Cruz-Escobar EN, Garibay-Díaz JC, León-Rodríguez E, De Jesús Ibarra-Sánchez M. Paclitaxel resistance is mediated by NF-κB on mesenchymal primary breast cancer cells. Oncol Lett 2022; 23:50. [PMID: 34992683 PMCID: PMC8721864 DOI: 10.3892/ol.2021.13168] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 11/09/2021] [Indexed: 12/16/2022] Open
Abstract
Paclitaxel has been used widely to treat breast cancer and other types of cancer. However, resistance is a major cause of failure for treatment and results in cancer progression. The present study investigated the association between paclitaxel resistance and the mesenchymal phenotype, using a model of primary breast cancer cells and employing four different cultures, two with an epithelial phenotype (MBCDF and MBCD17) and two with a mesenchymal phenotype (MBCDF-D5 and MBCD3). Epithelial-mesenchymal markers were evaluated by western blotting; MBCDF and MBCD17 cells expressed E-cadherin, SNAIL, Slug, and Twist, low levels of N-cadherin, but not vimentin. MBCDF-D5 and MBCD3 cells expressed N-cadherin, vimentin, and higher levels of SNAIL, and low levels of E-cadherin, Slug, and Twist. Cell viability was evaluated using a crystal violet assay after paclitaxel treatment; primary breast cancer cells with mesenchymal phenotype were resistant to paclitaxel compared with the epithelial primary breast cancer cells. Furthermore, using western blotting, it was revealed that mesenchymal cells had elevated levels of nuclear factor-κΒ (NF-κB) p65 and IκB kinase (IKK). Additionally, it was demonstrated that paclitaxel-induced degradation of the inhibitor of NF-κB, activation of NF-κB in a dose-dependent manner, and Bcl-2 and Bcl-xL upregulation. Finally, employing western blotting and crystal violet assays, the effects of the proteasome inhibitor ALLN were assessed. ALLN inhibited paclitaxel-induced NF-κB activation and restored the sensitivity to paclitaxel. Together, these data suggest that targeting the NF-κB/IKK axis might be a promising strategy to overcome paclitaxel resistance.
Collapse
Affiliation(s)
- José Esparza-López
- Biochemistry Unit, Salvador Zubirán National Institute of Health Sciences and Nutrition, Mexico City 14080, Mexico.,Research Support Network, National Autonomous University of Mexico-Salvador Zubirán National Institute of Health Sciences and Nutrition, Mexico City 14080, Mexico
| | - Ossian Longoria
- Hematology and Oncology Department, Salvador Zubirán National Institute of Health Sciences and Nutrition, Mexico City 14080, Mexico
| | | | - Julio Cesar Garibay-Díaz
- Hematology and Oncology Department, Salvador Zubirán National Institute of Health Sciences and Nutrition, Mexico City 14080, Mexico
| | - Eucario León-Rodríguez
- Hematology and Oncology Department, Salvador Zubirán National Institute of Health Sciences and Nutrition, Mexico City 14080, Mexico
| | | |
Collapse
|
6
|
Elias R, Tcheuyap VT, Kaushik AK, Singla N, Gao M, Reig Torras O, Christie A, Mulgaonkar A, Woolford L, Stevens C, Kettimuthu KP, Pavia-Jimenez A, Boroughs LK, Joyce A, Dakanali M, Notgrass H, Margulis V, Cadeddu JA, Pedrosa I, Williams NS, Sun X, DeBerardinis RJ, Öz OK, Zhong H, Seshagiri S, Modrusan Z, Cantarel BL, Kapur P, Brugarolas J. A renal cell carcinoma tumorgraft platform to advance precision medicine. Cell Rep 2021; 37:110055. [PMID: 34818533 PMCID: PMC8762721 DOI: 10.1016/j.celrep.2021.110055] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/10/2021] [Accepted: 11/03/2021] [Indexed: 12/30/2022] Open
Abstract
Renal cell carcinoma (RCC) encompasses a heterogenous group of tumors, but representative preclinical models are lacking. We previously showed that patient-derived tumorgraft (TG) models recapitulate the biology and treatment responsiveness. Through systematic orthotopic implantation of tumor samples from 926 ethnically diverse individuals into non-obese diabetic (NOD)/severe combined immunodeficiency (SCID) mice, we generate a resource comprising 172 independently derived, stably engrafted TG lines from 148 individuals. TG lines are characterized histologically and genomically (whole-exome [n = 97] and RNA [n = 102] sequencing). The platform features a variety of histological and oncogenotypes, including TCGA clades further corroborated through orthogonal metabolomic analyses. We illustrate how it enables a deeper understanding of RCC biology; enables the development of tissue- and imaging-based molecular probes; and supports advances in drug development.
Collapse
Affiliation(s)
- Roy Elias
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vanina T Tcheuyap
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Akash K Kaushik
- Howard Hughes Medical Institute and Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nirmish Singla
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Urology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ming Gao
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Oscar Reig Torras
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alana Christie
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Division of Biostatistics, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Aditi Mulgaonkar
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Layton Woolford
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christina Stevens
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kavitha Priya Kettimuthu
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrea Pavia-Jimenez
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lindsey K Boroughs
- Howard Hughes Medical Institute and Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Allison Joyce
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marianna Dakanali
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hollis Notgrass
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vitaly Margulis
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Urology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey A Cadeddu
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Urology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ivan Pedrosa
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Noelle S Williams
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiankai Sun
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ralph J DeBerardinis
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Howard Hughes Medical Institute and Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Orhan K Öz
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hua Zhong
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Somasekar Seshagiri
- Department of Microchemistry, Proteomics, Lipidomics and NGS, Genentech, Inc., South San Francisco, CA, USA
| | - Zora Modrusan
- Department of Microchemistry, Proteomics, Lipidomics and NGS, Genentech, Inc., South San Francisco, CA, USA
| | - Brandi L Cantarel
- Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Payal Kapur
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Urology, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
7
|
Kiang KMY, Chan AA, Leung GKK. Secondary gliosarcoma: the clinicopathological features and the development of a patient-derived xenograft model of gliosarcoma. BMC Cancer 2021; 21:265. [PMID: 33706745 PMCID: PMC7948380 DOI: 10.1186/s12885-021-08008-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/02/2021] [Indexed: 12/03/2022] Open
Abstract
Background Gliosarcoma (GSM) is a distinct and aggressive variant of glioblastoma multiforme (GBM) with worse prognosis and few treatment options. It is often managed with the same treatment modalities with temozolomide (TMZ) as in GBM. However, the therapeutic benefits on GSM from such treatment regimen is largely unknown. Patient-derived xenograft (PDX) models have been used widely to model tumor progression, and subsequently to validate biomarkers and inform potential therapeutic regimens. Here, we report for the first time the successful development of a PDX model of secondary GSM. Methods Tissue obtained from a tumor resection revealed a secondary GSM arising from GBM. The clinical, radiological, and histopathological records of the patient were retrospectively reviewed. Samples obtained from surgery were cultured ex vivo and/or implanted subcutaneously in immunocompromised mice. Histopathological features between the primary GBM, secondary GSM, and GSM PDX are compared. Results In explant culture, the cells displayed a spindle-shaped morphology under phase contrast microscopy, consistent with the sarcomatous component. GSM samples were subcutaneously engrafted into immunocompromised mice after single-cell suspension. Xenografts of serial passages showed enhanced growth rate with increased in vivo passage. We did not observe any histopathological differences between the secondary GSM and its serial in vivo passages of PDX tumors. Conclusions Our PDX model for GSM retained the histopathological characteristics of the engrafted tumor from the patient. It may provide valuable information to facilitate molecular and histopathological modelling of GSM and be of significant implication in future research to establish precise cancer medicine for this highly malignant tumor.
Collapse
Affiliation(s)
- Karrie Mei-Yee Kiang
- Division of Neurosurgery, Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, China
| | - Andrian A Chan
- Division of Neurosurgery, Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, China
| | - Gilberto Ka-Kit Leung
- Division of Neurosurgery, Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, China.
| |
Collapse
|
8
|
Abstract
The prognosis for childhood cancer has improved considerably over the past 50 years. This improvement is attributed to well-designed clinical trials which have incorporated chemotherapy, surgery, and radiation. With an increased understanding of cancer biology and genetics, we have entered an era of precision medicine and immunotherapy that provides potential for improved cure rates. However, preclinical evaluation of these therapies is more nuanced, requiring more robust animal models. Evaluation of targeted treatments requires molecularly defined xenograft models that can capture the diversity within pediatric cancer. The development of novel immunotherapies ideally involves the use of animal models that can accurately recapitulate the human immune response. In this review, we provide an overview of xenograft models for childhood cancers, review successful examples of novel therapies translated from xenograft models to the clinic, and describe the modern tools of xenograft biobanks and humanized xenograft models for the study of immunotherapies.
Collapse
Affiliation(s)
- Kevin O McNerney
- Children’s Hospital of Philadelphia, Divisions of Hematology and Oncology, Philadelphia, PA 19104, USA
| | - David T Teachey
- Children’s Hospital of Philadelphia, Divisions of Hematology and Oncology, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
9
|
Tian H, Lyu Y, Yang YG, Hu Z. Humanized Rodent Models for Cancer Research. Front Oncol 2020; 10:1696. [PMID: 33042811 PMCID: PMC7518015 DOI: 10.3389/fonc.2020.01696] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/30/2020] [Indexed: 12/18/2022] Open
Abstract
As one of the most popular laboratory animal models, rodents have been playing crucial roles in mechanistic investigations of oncogenesis as well as anticancer drug or regimen discoveries. However, rodent tumors show different or no responses to therapies against human cancers, and thus, in recent years, increased attention has been given to mouse models with xenografted or spontaneous human cancer cells. By combining with the human immune system (HIS) mice, these models have become more sophisticated and robust, enabling in vivo exploration of human cancer immunology and immunotherapy. In this review, we summarize the pros and cons of these humanized mouse models, with a focus on their potential as an in vivo platform for human cancer research. We also discuss the strategies for further improving these models.
Collapse
Affiliation(s)
- Huimin Tian
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Yanan Lyu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China.,International Center of Future Science, Jilin University, Changchun, China
| | - Zheng Hu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| |
Collapse
|
10
|
Modeling clear cell renal cell carcinoma and therapeutic implications. Oncogene 2020; 39:3413-3426. [PMID: 32123314 PMCID: PMC7194123 DOI: 10.1038/s41388-020-1234-3] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/12/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023]
Abstract
Renal cell carcinoma (RCC) comprises a diverse group of malignancies arising from the nephron. The most prevalent type, clear cell renal cell carcinoma (ccRCC), is characterized by genetic mutations in factors governing the hypoxia signaling pathway, resulting in metabolic dysregulation, heightened angiogenesis, intratumoral heterogeneity, and deleterious tumor microenvironmental (TME) crosstalk. Identification of specific genetic variances has led to therapeutic innovation and improved survival for patients with ccRCC. Current barriers to effective long-term therapeutic success highlight the need for continued drug development using improved modeling systems. ccRCC preclinical models can be grouped into three broad categories: cell line, mouse, and 3D models. Yet, the breadth of important unanswered questions in ccRCC research far exceeds the accessibility of model systems capable of carrying them out. Accordingly, we review the strengths, weaknesses, and therapeutic implications of each model system that are relied upon today.
Collapse
|
11
|
Sobczuk P, Brodziak A, Khan MI, Chhabra S, Fiedorowicz M, Wełniak-Kamińska M, Synoradzki K, Bartnik E, Cudnoch-Jędrzejewska A, Czarnecka AM. Choosing The Right Animal Model for Renal Cancer Research. Transl Oncol 2020; 13:100745. [PMID: 32092671 PMCID: PMC7036425 DOI: 10.1016/j.tranon.2020.100745] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/04/2020] [Accepted: 01/06/2020] [Indexed: 12/17/2022] Open
Abstract
The increase in the life expectancy of patients with renal cell carcinoma (RCC) in the last decade is due to changes that have occurred in the area of preclinical studies. Understanding cancer pathophysiology and the emergence of new therapeutic options, including immunotherapy, would not be possible without proper research. Before new approaches to disease treatment are developed and introduced into clinical practice they must be preceded by preclinical tests, in which animal studies play a significant role. This review describes the progress in animal model development in kidney cancer research starting from the oldest syngeneic or chemically-induced models, through genetically modified mice, finally to xenograft, especially patient-derived, avatar and humanized mouse models. As there are a number of subtypes of RCC, our aim is to help to choose the right animal model for a particular kidney cancer subtype. The data on genetic backgrounds, biochemical parameters, histology, different stages of carcinogenesis and metastasis in various animal models of RCC as well as their translational relevance are summarized. Moreover, we shed some light on imaging methods, which can help define tumor microstructure, assist in the analysis of its metabolic changes and track metastasis development.
Collapse
Affiliation(s)
- Paweł Sobczuk
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland; Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland.
| | - Anna Brodziak
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland; Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland.
| | - Mohammed Imran Khan
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada.
| | - Stuti Chhabra
- Department of Biochemistry, CSIR-Central Drug Research Institute, Lucknow, India.
| | - Michał Fiedorowicz
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| | - Marlena Wełniak-Kamińska
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| | - Kamil Synoradzki
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| | - Ewa Bartnik
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| | - Anna M Czarnecka
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| |
Collapse
|
12
|
Tracey AT, Murray KS, Coleman JA, Kim K. Patient-Derived Xenograft Models in Urological Malignancies: Urothelial Cell Carcinoma and Renal Cell Carcinoma. Cancers (Basel) 2020; 12:cancers12020439. [PMID: 32069881 PMCID: PMC7072311 DOI: 10.3390/cancers12020439] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 12/17/2022] Open
Abstract
The engraftment of human tumor tissues into immunodeficient host mice to generate patient-derived xenograft (PDX) models has become increasingly utilized for many types of cancers. By capturing the unique genomic and molecular properties of the parental tumor, PDX models enable analysis of patient-specific clinical responses. PDX models are an important platform to address the contribution of inter-tumoral heterogeneity to therapeutic sensitivity, tumor evolution, and the mechanisms of treatment resistance. With the increasingly important role played by targeted therapies in urological malignancies, the establishment of representative PDX models can contribute to improved facilitation and adoption of precision medicine. In this review of the evolving role of the PDX in urothelial cancer and kidney cancer, we discuss the essential elements of successful graft development, effective translational application, and future directions for clinical models.
Collapse
Affiliation(s)
- Andrew T. Tracey
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (A.T.T.); (J.A.C.)
| | - Katie S. Murray
- Department of Surgery, Division of Urology, University of Missouri, Columbia, MO 65211, USA;
| | - Jonathan A. Coleman
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (A.T.T.); (J.A.C.)
| | - Kwanghee Kim
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Correspondence: ; Tel.: +1-646-422-4432
| |
Collapse
|
13
|
Patel A, Cohen S, Moret R, Maresh G, Gobe GC, Li L. Patient-derived xenograft models to optimize kidney cancer therapies. Transl Androl Urol 2019; 8:S156-S165. [PMID: 31236333 DOI: 10.21037/tau.2018.11.04] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Renal cell carcinoma (RCC) is the most common solid neoplasm of the adult kidney and has a high potential for developing metastatic spread. Approximately 25-30% of RCC patients have metastatic disease at presentation, and 30-40% of patients develop metastases after the initial diagnosis. Advanced renal cancer is a deadly and difficult-to-treat cancer. The 5-year survival rate of patients with metastatic disease is less than 10%, partly because RCC metastases become resistant to current therapies. Pre-clinical models may help to identify the optimum therapeutic options for individual patients. Here we reviewed various mouse xenograft methods for RCC treatment screening especially patient-derived orthotopic xenograft models. Advantages and disadvantaged of some of the models are also discussed.
Collapse
Affiliation(s)
- Avi Patel
- UQ-Ochsner Clinical School, Institute for Translational Research, Ochsner Clinic Foundation, New Orleans, LA, USA
| | - Sarah Cohen
- UQ-Ochsner Clinical School, Institute for Translational Research, Ochsner Clinic Foundation, New Orleans, LA, USA
| | - Ravan Moret
- UQ-Ochsner Clinical School, Institute for Translational Research, Ochsner Clinic Foundation, New Orleans, LA, USA
| | - Grace Maresh
- UQ-Ochsner Clinical School, Institute for Translational Research, Ochsner Clinic Foundation, New Orleans, LA, USA
| | - Glenda C Gobe
- UQ NHMRC CKD.QLD CRE, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia.,University of Queensland Princess Alexandra Hospital Kidney Disease Research Collaborative, Translational Research Institute, Brisbane, QLD, Australia
| | - Li Li
- UQ-Ochsner Clinical School, Institute for Translational Research, Ochsner Clinic Foundation, New Orleans, LA, USA
| |
Collapse
|
14
|
Experimental imaging in orthotopic renal cell carcinoma xenograft models: comparative evaluation of high-resolution 3D ultrasonography, in-vivo micro-CT and 9.4T MRI. Sci Rep 2017; 7:14249. [PMID: 29079842 PMCID: PMC5660163 DOI: 10.1038/s41598-017-14759-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/17/2017] [Indexed: 12/28/2022] Open
Abstract
In this study, we aimed to comparatively evaluate high-resolution 3D ultrasonography (hrUS), in-vivo micro-CT (μCT) and 9.4T MRI for the monitoring of tumor growth in an orthotopic renal cell carcinoma (RCC) xenograft model since there is a lack of validated, non-invasive imaging tools for this purpose. 1 × 106 Caki-2 RCC cells were implanted under the renal capsule of 16 immunodeficient mice. Local and systemic tumor growth were monitored by regular hrUS, μCT and MRI examinations. Cells engrafted in all mice and gave rise to exponentially growing, solid tumors. All imaging techniques allowed to detect orthotopic tumors and to precisely calculate their volumes. While tumors appeared homogenously radiolucent in μCT, hrUS and MRI allowed for a better visualization of intratumoral structures and surrounding soft tissue. Examination time was the shortest for hrUS, followed by μCT and MRI. Tumor volumes determined by hrUS, μCT and MRI showed a very good correlation with each other and with caliper measurements at autopsy. 10 animals developed pulmonary metastases being well detectable by μCT and MRI. In conclusion, each technique has specific strengths and weaknesses, so the one(s) best suitable for a specific experiment may be chosen individually.
Collapse
|
15
|
Shi H, Yu J, Li J. Nephroblastoma overexpressed gene expression and its prognostic implications of clinical outcomes in renal cell carcinoma patients. Cancer Biomark 2017; 20:241-246. [PMID: 28800310 DOI: 10.3233/cbm-170017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The nephroblastoma overexpressed gene (NOV) expressions in tissues and organs has become abnormal during tumorigenesis and progression. This study intended to investigate the correlation between clinical outcomes and NOV expression in renal cell carcinoma (RCC) patients. METHODS Fifty RCC patients who attended the hospital from January 2013 to January 2015 were enrolled in this study. NOV expression in cancerous tissues and adjacent non-tumor (ANT) renal tissues of RCC patients was detected by immunohistochemistry (IHC). According to the percentage of NOV-positive cells, cases were divided into NOV-positive and NOV-negative groups. The correlations between age, gender, disease course, tumor diameter, pathological grades (WHO/ISUP grading system) or tumor-node-metastasis (TNM) staging and NOV-positive rate were determined. Kaplan-Meier method was utilized for analyzing the 3- and 5-survial rates of RCC patients. The Cox proportional hazards regression model was used for the multivariate analysis. RESULTS NOV-positive rate was uncorrelated with age, gender, disease course or TNM classification while was negatively correlated with pathological grades. NOV-positive rate in RCC tumor and ANT tissues was 58% and 100%, respectively. Five-year survival rate in NOV-positive group was significantly lower than that in NOV-negative group. CONCLUSION Our data suggested that NOV down-regulation might be a biomarker for RCC but its positivity might be an indicator of poor prognosis.
Collapse
Affiliation(s)
- Hongbin Shi
- Department of Urology Surgery, Ningxia People's Hospital, Yinchuan 750001, Ningxia, China
| | - Jianping Yu
- Department of Laboratory, Xianyang Hospital of Yan'an University, Xianyang 712000, Shaanxi, China
| | - Jie Li
- Department of Nephropathy, Xi'an Central Hospital, Xi'an 710003, Shaanxi, China
| |
Collapse
|
16
|
Inoue T, Terada N, Kobayashi T, Ogawa O. Patient-derived xenografts as in vivo models for research in urological malignancies. Nat Rev Urol 2017; 14:267-283. [PMID: 28248952 DOI: 10.1038/nrurol.2017.19] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lack of appropriate models that recapitulate the complexity and heterogeneity of urological tumours precludes most of the preclinical reagents that target urological tumours from receiving regulatory approval. Patient-derived xenograft (PDX) models are characterized by direct engraftment of patient-derived tumour fragments into immunocompromised mice. PDXs can maintain the original histology, as well as the molecular and genetic characteristics of the source tumour. Thus, PDX models have various advantages over conventional cell-line-derived xenograft (CDX) and other models, which has resulted in an increase in the use of urological tumour PDXs in the analysis of tumour biology and, importantly, for drug development and treatment decisions in personalized medicine. PDX models of urological malignancies have great potential to be used for both basic and clinical research, but limitations exist and need to be overcome. In particular, several agents targeting the immune system have shown promising results in kidney and bladder cancer; however, establishing PDX models in mice with an intact immune system so that an immune response against the tumour is triggered is important to investigate these new therapeutics. Moreover, international collaboration to share PDX models is essential for research concerning fatal urological tumours.
Collapse
Affiliation(s)
- Takahiro Inoue
- Department of Urology, Kyoto University Graduate School of Medicine, 54 Kawaharacho Shogoin Sakyo-ku, Kyoto, 6068507, Japan
| | - Naoki Terada
- Department of Urology, Kyoto University Graduate School of Medicine, 54 Kawaharacho Shogoin Sakyo-ku, Kyoto, 6068507, Japan
| | - Takashi Kobayashi
- Department of Urology, Kyoto University Graduate School of Medicine, 54 Kawaharacho Shogoin Sakyo-ku, Kyoto, 6068507, Japan
| | - Osamu Ogawa
- Department of Urology, Kyoto University Graduate School of Medicine, 54 Kawaharacho Shogoin Sakyo-ku, Kyoto, 6068507, Japan
| |
Collapse
|
17
|
Shologu N, Szegezdi E, Lowery A, Kerin M, Pandit A, Zeugolis DI. Recreating complex pathophysiologies in vitro with extracellular matrix surrogates for anticancer therapeutics screening. Drug Discov Today 2016; 21:1521-1531. [DOI: 10.1016/j.drudis.2016.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 05/17/2016] [Accepted: 06/01/2016] [Indexed: 12/12/2022]
|
18
|
Cancer stem cells are underestimated by standard experimental methods in clear cell renal cell carcinoma. Sci Rep 2016; 6:25220. [PMID: 27121191 PMCID: PMC4848484 DOI: 10.1038/srep25220] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/13/2016] [Indexed: 01/06/2023] Open
Abstract
Rare cancer stem cells (CSC) are proposed to be responsible for tumour propagation and re-initiation and are functionally defined by identifying tumour-initiating cells (TICs) using the xenotransplantation limiting dilution assay (LDA). While TICs in clear cell renal cell carcinoma (ccRCC) appeared rare in NOD/SCID/IL2Rγ(-/-) (NSG) mice, xenografts formed more efficiently from small tumour fragments, indicating the LDA underestimated ccRCC TIC frequency. Mechanistic interrogation of the LDA identified multiple steps that influence ccRCC TIC quantitation. For example, tissue disaggregation destroys most ccRCC cells, common assays significantly overestimate tumour cell viability, and microenvironmental supplementation with human extracellular factors or pharmacological inhibition of anoikis increase clonogenicity and tumourigenicity of ccRCC cell lines and primary tumour cells. Identification of these previously uncharacterized concerns that cumulatively lead to substantial underestimation of TICs in ccRCC provides a framework for development of more accurate TIC assays in the future, both for this disease and for other cancers.
Collapse
|
19
|
Porta C, Giglione P, Liguigli W, Paglino C. Dovitinib (CHIR258, TKI258): structure, development and preclinical and clinical activity. Future Oncol 2015; 11:39-50. [PMID: 25572783 DOI: 10.2217/fon.14.208] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Dovitinib is an oral multikinase inhibitor targeting FGF receptors, PDGF receptors and VEGF receptors. Its activity against FGF receptors suggests its usefulness in treating cancers after the failure of VEGF/VEGF receptor-targeting agents. The identified dose and schedule to be used in further studies was 500 mg orally for 5 days on and 2 days off. Biological considerations and the results achieved in a Phase I/II trial suggested its activity in advanced renal cell carcinoma patients pretreated with a tyrosine kinase inhibitor and an mTOR inhibitor. Surprisingly, in a randomized controlled Phase III trial versus sorafenib in the same setting, dovitinib failed to demonstrate any superiority. At present, dovitinib is being tested in different tumor types. However, molecular-based patient selection seems to be key to fully exploit the activity of this drug.
Collapse
Affiliation(s)
- Camillo Porta
- Medical Oncology, IRCCS San Matteo University Hospital Foundation, Piazzale C Golgi, 19, 27100 Pavia, Italy
| | | | | | | |
Collapse
|
20
|
Chang DK, Moniz RJ, Xu Z, Sun J, Signoretti S, Zhu Q, Marasco WA. Human anti-CAIX antibodies mediate immune cell inhibition of renal cell carcinoma in vitro and in a humanized mouse model in vivo. Mol Cancer 2015; 14:119. [PMID: 26062742 PMCID: PMC4464115 DOI: 10.1186/s12943-015-0384-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/11/2015] [Indexed: 12/17/2022] Open
Abstract
Background Carbonic anhydrase (CA) IX is a surface-expressed protein that is upregulated by the hypoxia inducible factor (HIF) and represents a prototypic tumor-associated antigen that is overexpressed on renal cell carcinoma (RCC). Therapeutic approaches targeting CAIX have focused on the development of CAIX inhibitors and specific immunotherapies including monoclonal antibodies (mAbs). However, current in vivo mouse models used to characterize the anti-tumor properties of fully human anti-CAIX mAbs have significant limitations since the role of human effector cells in tumor cell killing in vivo is not directly evaluated. Methods The role of human anti-CAIX mAbs on CAIX+ RCC tumor cell killing by immunocytes or complement was tested in vitro by antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC) and antibody-dependent cellular phagocytosis (ADCP) as well as on CAIX+ RCC cellular motility, wound healing, migration and proliferation. The in vivo therapeutic activity mediated by anti-CAIX mAbs was determined by using a novel orthotopic RCC xenograft humanized animal model and analyzed by histology and FACS staining. Results Our studies demonstrate the capacity of human anti-CAIX mAbs that inhibit CA enzymatic activity to result in immune-mediated killing of RCC, including nature killer (NK) cell-mediated ADCC, CDC, and macrophage-mediated ADCP. The killing activity correlated positively with the level of CAIX expression on RCC tumor cell lines. In addition, Fc engineering of anti-CAIX mAbs was shown to enhance the ADCC activity against RCC. We also demonstrate that these anti-CAIX mAbs inhibit migration of RCC cells in vitro. Finally, through the implementation of a novel orthotopic RCC model utilizing allogeneic human peripheral blood mononuclear cells in NOD/SCID/IL2Rγ−/− mice, we show that anti-CAIX mAbs are capable of mediating human immune response in vivo including tumor infiltration of NK cells and activation of T cells, resulting in inhibition of CAIX+ tumor growth. Conclusions Our findings demonstrate that these novel human anti-CAIX mAbs have therapeutic potential in the unmet medical need of targeted killing of HIF-driven CAIX+RCC. The orthotopic tumor xenografted humanized mouse provides an improved model to evaluate the in vivo anti-tumor capabilities of fully human mAbs for RCC therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0384-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- De-Kuan Chang
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| | - Raymond J Moniz
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| | - Zhongyao Xu
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA.
| | - Jiusong Sun
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| | - Sabina Signoretti
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Quan Zhu
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| | - Wayne A Marasco
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| |
Collapse
|
21
|
Establishment and characterization of patient-derived tumor xenograft using gastroscopic biopsies in gastric cancer. Sci Rep 2015; 5:8542. [PMID: 25712750 PMCID: PMC4339807 DOI: 10.1038/srep08542] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 01/23/2015] [Indexed: 12/29/2022] Open
Abstract
The patient-derived tumor xenograft (PDTX) model has become the most realistic model for preclinical studies. PDTX models of gastric cancer using surgical tissues are reported occasionally; however, the PDTX models using gastroscopic biopsies, which are best for evaluating new drugs, are unreported. In our study, a total of 185 fresh gastroscopic biopsies of gastric cancer were subcutaneously transplanted into NOD/SCID (Nonobese Diabetic/Severe Combined Immunodeficiency) mice. Sixty-three PDTX models were successfully established (34.1%, 63/185) and passaged to maintain tumors in vivo, and the mean latency period of xenografts was 65.86 ± 32.84 days (11–160 days). Biopsies of prior chemotherapy had a higher transplantation rate (52.1%, 37/71) than biopsies after chemotherapy (21.9%, 25/114; P = 0.000). No differences were found between the latency period of xenografts and characteristics of patients. The pathological and molecular features of PDTX as well as chemosensitivity were highly consistent with those of primary tumors of patients. The genetic characteristics were stable during passaging of PDTX models. In summary PDTX models using gastroscopic biopsies in gastric cancer were demonstrated for the first time, and the biological characteristics of the PDTX models were highly consistent with patients, which provided the best preclinical study platform for gastric cancer.
Collapse
|
22
|
Varna M, Bertheau P, Legrès L. Tumor Microenvironment in Human Tumor Xenografted Mouse Models. ACTA ACUST UNITED AC 2014. [DOI: 10.6000/1927-7229.2014.03.03.6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
23
|
Pavía-Jiménez A, Tcheuyap VT, Brugarolas J. Establishing a human renal cell carcinoma tumorgraft platform for preclinical drug testing. Nat Protoc 2014; 9:1848-59. [PMID: 25010905 DOI: 10.1038/nprot.2014.108] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Traditionally, xenograft models have been used to study tumors in vivo. However, their utility is reduced by the use of tumor cell lines for implantation. Tumorgrafts (TGs; also known as patient-derived xenografts (PDXs)), which involve patient-derived tumor samples, are increasingly recognized as more representative models than traditional xenografts. Furthermore, we showed previously that renal cell carcinoma (RCC) TGs retain the histology, gene expression, DNA copy number alterations, mutations and treatment responsiveness of patient tumors. In skilled hands, implantations require ≤5 min per mouse, and TGs typically grow to 1 cm in 1-4 months. Here we outline the process of implantation of patient-derived RCC samples into the kidneys of immunodeficient mice, as well as the s.c. implantation for preclinical drug testing, including guidelines for the design and execution of drug trials. TGs have extensive applications besides therapeutic studies and may identify biomarkers and mechanisms of resistance. In addition, they may provide insights into tumor biology.
Collapse
Affiliation(s)
- Andrea Pavía-Jiménez
- 1] Kidney Cancer Program, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA. [2] Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA. [3] Department of Developmental Biology, UT Southwestern Medical Center, Dallas, Texas, USA. [4] Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Vanina Toffessi Tcheuyap
- 1] Kidney Cancer Program, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA. [2] Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA. [3] Department of Developmental Biology, UT Southwestern Medical Center, Dallas, Texas, USA. [4] Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas, USA
| | - James Brugarolas
- 1] Kidney Cancer Program, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA. [2] Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA. [3] Department of Developmental Biology, UT Southwestern Medical Center, Dallas, Texas, USA. [4] Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
24
|
Varna M, Bousquet G, Ferreira I, Goulard M, El-Bouchtaoui M, Artus PM, Verine J, de Kerviler E, Hernandez L, Leboeuf C, Escudier B, Legrès L, Setterblad N, Soliman H, Feugeas JP, Janin A, Bertheau P. Stability of preclinical models of aggressive renal cell carcinomas. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:2950-2962. [PMID: 25031714 PMCID: PMC4097235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/25/2014] [Indexed: 06/03/2023]
Abstract
Renal-cell carcinomas (RCC) are often resistant to conventional cytotoxic agents. Xenograft models are used for in vivo preclinical studies and drug development. The validity of these studies is highly dependent on the phenotypic and genotypic stability of the models. Here we assessed the stability of six aggressive human RCC xenografted in nude/NMRI mice. We compared the initial samples (P0), first (P1) and fifth (P5) passages for the following criteria: histopathology, immunohistochemistry for CK7, CD10, vimentin and p53, DNA allelic profiles using 10 microsatellites and CGH-array. Next we evaluated the response to sunitinib in primary RCC and corresponding xenografted RCC. We observed a good overall stability between primary RCC and corresponding xenografted RCC at P1 and P5 regarding histopathology and immunohistochemistry except for cytokeratin 7 (one case) and p53 (one case) expression. Out of 44 groups with fully available microsatellite data (at P0, P1 and P5), 66% (29 groups) showed no difference from P0 to P5 while 34% (15 groups) showed new or lost alleles. Using CGH-array, overall genomic alterations at P5 were not different from those of initial RCC. The xenografted RCC had identical response to sunitinib therapy compared to the initial human RCC from which they derive. These xenograft models of aggressive human RCC are clinically relevant, showing a good histological and molecular stability and are suitable for studies of basic biology and response to therapy.
Collapse
Affiliation(s)
- Mariana Varna
- Université Paris Diderot, Sorbonne Paris CitéF-75010 Paris, France
- INSERM, U1165Paris, F-75010 France
- AP-HP-Hôpital Saint-Louis, Laboratoire de PathologieParis, F-75010 France
| | - Guilhem Bousquet
- Université Paris Diderot, Sorbonne Paris CitéF-75010 Paris, France
- INSERM, U1165Paris, F-75010 France
- AP-HP-Hôpital Saint-Louis, Laboratoire de PathologieParis, F-75010 France
| | - Irmine Ferreira
- Université Paris Diderot, Sorbonne Paris CitéF-75010 Paris, France
- INSERM, U1165Paris, F-75010 France
- AP-HP-Hôpital Saint-Louis, Laboratoire de PathologieParis, F-75010 France
| | - Marie Goulard
- Université Paris Diderot, Sorbonne Paris CitéF-75010 Paris, France
- INSERM, U1165Paris, F-75010 France
| | - Morad El-Bouchtaoui
- Université Paris Diderot, Sorbonne Paris CitéF-75010 Paris, France
- INSERM, U1165Paris, F-75010 France
- AP-HP-Hôpital Saint-Louis, Laboratoire de PathologieParis, F-75010 France
| | - Pierre Mongiat Artus
- Université Paris Diderot, Sorbonne Paris CitéF-75010 Paris, France
- INSERM, U1165Paris, F-75010 France
- AP-HP-Hôpital Saint-Louis, Service d’UrologieParis, F-75010 France
| | - Jérome Verine
- Université Paris Diderot, Sorbonne Paris CitéF-75010 Paris, France
- INSERM, U1165Paris, F-75010 France
- AP-HP-Hôpital Saint-Louis, Laboratoire de PathologieParis, F-75010 France
| | - Eric de Kerviler
- Université Paris Diderot, Sorbonne Paris CitéF-75010 Paris, France
- INSERM, U1165Paris, F-75010 France
- AP-HP-Hôpital Saint-Louis, Service de RadiologieParis, F-75010 France
| | - Lucie Hernandez
- Université Paris Diderot, Sorbonne Paris CitéF-75010 Paris, France
| | - Christophe Leboeuf
- Université Paris Diderot, Sorbonne Paris CitéF-75010 Paris, France
- INSERM, U1165Paris, F-75010 France
- AP-HP-Hôpital Saint-Louis, Laboratoire de PathologieParis, F-75010 France
| | | | - Luc Legrès
- Université Paris Diderot, Sorbonne Paris CitéF-75010 Paris, France
- INSERM, U1165Paris, F-75010 France
| | | | - Hany Soliman
- AP-HP-Hôpital Saint-Louis, Service de BiochimieParis, F-75010 France
| | - Jean-Paul Feugeas
- AP-HP-Hôpital Saint-Louis, Service de BiochimieParis, F-75010 France
| | - Anne Janin
- Université Paris Diderot, Sorbonne Paris CitéF-75010 Paris, France
- INSERM, U1165Paris, F-75010 France
- AP-HP-Hôpital Saint-Louis, Laboratoire de PathologieParis, F-75010 France
| | - Philippe Bertheau
- Université Paris Diderot, Sorbonne Paris CitéF-75010 Paris, France
- INSERM, U1165Paris, F-75010 France
- AP-HP-Hôpital Saint-Louis, Laboratoire de PathologieParis, F-75010 France
| |
Collapse
|
25
|
Valta MP, Zhao H, Ingels A, Thong AE, Nolley R, Saar M, Peehl DM. Development of a realistic in vivo bone metastasis model of human renal cell carcinoma. Clin Exp Metastasis 2014; 31:573-84. [PMID: 24715498 DOI: 10.1007/s10585-014-9651-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 03/19/2014] [Indexed: 02/06/2023]
Abstract
About one-third of patients with advanced renal cell carcinoma (RCC) have bone metastases. The incidence of RCC is increasing and bone metastatic RCC merits greater focus. Realistic preclinical bone metastasis models of RCC are lacking, hampering the development of effective therapies. We developed a realistic in vivo bone metastasis model of human RCC by implanting precision-cut tissue slices under the renal capsule of immunodeficient mice. The presence of disseminated cells in bone marrow of tissue slice graft (TSG)-bearing mice was screened by human-specific polymerase chain reaction and confirmed by immunohistology using human-specific antibody. Disseminated tumor cells in bone marrow of TSG-bearing mice derived from three of seven RCC patients were detected as early as 1 month after tissue implantation at a high frequency with close resemblance to parent tumors (e.g., CAIX expression and high vascularity). The metastatic patterns of TSGs correlated with disease progression in patients. In addition, TSGs retained capacity to metastasize to bone at high frequency after serial passaging and cryopreservation. Moreover, bone metastases in mice responded to Temsirolimus treatment. Intratibial injections of single cells generated from TSGs showed 100 % engraftment and produced X-ray-visible tumors as early as 3 weeks after cancer cell inoculation. Micro-computed tomography (μCT) and histological analysis revealed osteolytic characteristics of these lesions. Our results demonstrated that orthotopic RCC TSGs have potential to develop bone metastases that respond to standard therapy. This first reported primary RCC bone metastasis model provides a realistic setting to test therapeutics to prevent or treat bone metastases in RCC.
Collapse
Affiliation(s)
- Maija P Valta
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Konstantinopoulos PA, Matulonis UA. Current status and evolution of preclinical drug development models of epithelial ovarian cancer. Front Oncol 2013; 3:296. [PMID: 24377084 PMCID: PMC3858677 DOI: 10.3389/fonc.2013.00296] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 11/23/2013] [Indexed: 01/06/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy and the fifth most common cause of female cancer death in the United States. Although important advances in surgical and chemotherapeutic strategies over the last three decades have significantly improved the median survival of EOC patients, the plateau of the survival curve has not changed appreciably. Given that EOC is a genetically and biologically heterogeneous disease, identification of specific molecular abnormalities that can be targeted in each individual ovarian cancer on the basis of predictive biomarkers promises to be an effective strategy to improve outcome in this disease. However, for this promise to materialize, appropriate preclinical experimental platforms that recapitulate the complexity of these neoplasms and reliably predict antitumor activity in the clinic are critically important. In this review, we will present the current status and evolution of preclinical models of EOC, including cell lines, immortalized normal cells, xenograft models, patient-derived xenografts, and animal models, and will discuss their potential for oncology drug development.
Collapse
Affiliation(s)
- Panagiotis A Konstantinopoulos
- Medical Gynecologic Oncology Program, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School , Boston, MA , USA
| | - Ursula A Matulonis
- Medical Gynecologic Oncology Program, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
27
|
Thong AE, Zhao H, Ingels A, Valta MP, Nolley R, Santos J, Young SR, Peehl DM. Tissue slice grafts of human renal cell carcinoma: an authentic preclinical model with high engraftment rate and metastatic potential. Urol Oncol 2013; 32:43.e23-30. [PMID: 23911681 DOI: 10.1016/j.urolonc.2013.05.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 05/14/2013] [Accepted: 05/15/2013] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Discovery of curative therapies for renal cell carcinoma (RCC) is hampered by lack of authentic preclinical models. Tumorgrafts, generated by direct implantation of patient-derived tissues into mice, have demonstrated superior ability to predict therapeutic response. We evaluated "tissue slice grafts" (TSGs) as an improved tumorgraft model of RCC. MATERIALS AND METHODS Cores of fresh RCC were precision-cut at 300 µm and implanted under the renal capsule of RAG2(-/-)γC(-/-) mice. Engraftment rate, histology, biomarker expression, genetic fidelity, and metastatic potential were evaluated. Magnetic resonance imaging (MRI) was tested as a noninvasive method to measure tumor volume, and response to a targeted therapy was determined. RESULTS All 13 cases of RCC engrafted and displayed characteristic histology and biomarkers. TSG volume quantified noninvasively by MRI highly correlated with graft weights, providing a unique tool for monitoring orthotopic growth. Moreover, in 2 cases, cancer cells from TSGs metastasized to clinically relevant sites, including bone. Microarray analysis and DNA sequencing demonstrated a high degree of correlation of global gene expression and von Hippel-Lindau (VHL) status between TSGs and parental tumors. Treatment of TSGs with sunitinib significantly decreased graft weight and mean vessel density compared with controls. CONCLUSION The TSG model of RCC faithfully recapitulates tumor pathology, gene expression, genetic mutation, and drug response. The high engraftment rate and metastatic potential of this authentic model, in conjunction with the ability to generate large first-generation animal cohorts and to quantitate tumor volume at the orthotopic site by MRI, proffer significant advantages compared with other preclinical platforms.
Collapse
Affiliation(s)
- Alan E Thong
- Department of Urology, Stanford University School of Medicine, Stanford, CA
| | - Hongjuan Zhao
- Department of Urology, Stanford University School of Medicine, Stanford, CA
| | - Alexandre Ingels
- Department of Urology, Stanford University School of Medicine, Stanford, CA; Department of Urology, Centre Hospitalier Universitaire du Kremlin-Bicêtre, Kremlin-Bicêtre, France
| | - Maija P Valta
- Department of Urology, Stanford University School of Medicine, Stanford, CA; Division of Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Rosalie Nolley
- Department of Urology, Stanford University School of Medicine, Stanford, CA
| | - Jennifer Santos
- Department of Urology, Stanford University School of Medicine, Stanford, CA
| | - Sarah R Young
- Department of Urology, Stanford University School of Medicine, Stanford, CA
| | - Donna M Peehl
- Department of Urology, Stanford University School of Medicine, Stanford, CA.
| |
Collapse
|
28
|
Origin of the vasculature supporting growth of primary patient tumor xenografts. J Transl Med 2013; 11:110. [PMID: 23639003 PMCID: PMC3660244 DOI: 10.1186/1479-5876-11-110] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Accepted: 04/29/2013] [Indexed: 01/11/2023] Open
Abstract
Background Studies of primary patient tumor xenografts grown in immunodeficient mice have shown that these tumors histologically and genetically closely resemble the original tumors. These patient xenograft models are becoming widely used for therapeutic efficacy studies. Because many therapies are directed at tumor stromal components and because the tumor microenvironment also is known to influence the response of a tumor to therapy, it is important to understand the nature of the stroma and, in particular, the vascular supply of patient xenografts. Methods Patient tumor xenografts were established by implanting undisrupted pieces of patient tumors in SCID mice. For this study, formalin fixed, paraffin embedded specimens from several types of solid tumors were selected and, using species-specific antibodies which react with formalin fixed antigens, we analyzed the species origin of the stroma and blood vessels that supported tumor growth in these models. Additionally, we investigated the kinetics of the vascularization process in a colon tumor and a mesothelioma xenograft. In mice bearing a head and neck xenograft, a perfusion study was performed to compare the functionality of the human and mouse tumor vessels. Results In patient tumors which successfully engrafted, the human stroma and vessels which were engrafted as part of the original tumor did not survive and were no longer detectable at the time of first passage (15–25 weeks). Uniformly, the stroma and vessels supporting the growth of these tumors were of murine origin. The results of the kinetic studies showed that the loss of the human vessels and vascularization by host vessels occurred more rapidly in a colon tumor (by 3 weeks) than in a mesothelioma (by 9 weeks). Finally, the perfusion studies revealed that while mouse vessels in the periphery of the tumor were perfused, those in the central regions were rarely perfused. No vessels of human origin were detected in this model. Conclusions In the tumors we investigated, we found no evidence that the human stromal cells and vessels contained in the original implant either survived or contributed in any substantive way to the growth of these xenografts.
Collapse
|
29
|
Sivanand S, Peña-Llopis S, Zhao H, Kucejova B, Spence P, Pavia-Jimenez A, Yamasaki T, McBride DJ, Gillen J, Wolff NC, Morlock L, Lotan Y, Raj GV, Sagalowsky A, Margulis V, Cadeddu JA, Ross MT, Bentley DR, Kabbani W, Xie XJ, Kapur P, Williams NS, Brugarolas J. A validated tumorgraft model reveals activity of dovitinib against renal cell carcinoma. Sci Transl Med 2012; 4:137ra75. [PMID: 22674553 DOI: 10.1126/scitranslmed.3003643] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Most anticancer drugs entering clinical trials fail to achieve approval from the U.S. Food and Drug Administration. Drug development is hampered by the lack of preclinical models with therapeutic predictive value. Herein, we report the development and validation of a tumorgraft model of renal cell carcinoma (RCC) and its application to the evaluation of an experimental drug. Tumor samples from 94 patients were implanted in the kidneys of mice without additives or disaggregation. Tumors from 35 of these patients formed tumorgrafts, and 16 stable lines were established. Samples from metastatic sites engrafted at higher frequency than those from primary tumors, and stable engraftment of primary tumors in mice correlated with decreased patient survival. Tumorgrafts retained the histology, gene expression, DNA copy number alterations, and more than 90% of the protein-coding gene mutations of the corresponding tumors. As determined by the induction of hypercalcemia in tumorgraft-bearing mice, tumorgrafts retained the ability to induce paraneoplastic syndromes. In studies simulating drug exposures in patients, RCC tumorgraft growth was inhibited by sunitinib and sirolimus (the active metabolite of temsirolimus in humans), but not by erlotinib, which was used as a control. Dovitinib, a drug in clinical development, showed greater activity than sunitinib and sirolimus. The routine incorporation of models recapitulating the molecular genetics and drug sensitivities of human tumors into preclinical programs has the potential to improve oncology drug development.
Collapse
Affiliation(s)
- Sharanya Sivanand
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Characterization of the Tumor-Microenvironment in Patient-Derived Cervix Xenografts (OCICx). Cancers (Basel) 2012; 4:821-45. [PMID: 24213469 PMCID: PMC3712708 DOI: 10.3390/cancers4030821] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 08/17/2012] [Accepted: 08/21/2012] [Indexed: 02/07/2023] Open
Abstract
Rationale: The tumor microenvironment (TME) is heterogeneous including both malignant and host cell components as well as regions of hypoxia, elevated interstitial fluid pressure (IFP) and poor nutrient supply. The quantitative extent to which the microenvironmental properties of primary tumors are recapitulated in xenograft models is not well characterized. Methods: Xenografts were generated by implanting tumor biopsies directly into the cervix of mice to create a panel of orthotopically-passaged xenografts (OCICx). Tumors were grown to ~1 cm (diameter) and IFP measurements recorded prior to sacrifice. Enlarged para-aortic lymph nodes (>1–2 mm) were excised for histologic confirmation of metastatic disease. Quantitative histological analysis was used to evaluate hypoxia, proliferation, lymphatic and blood vessels in the epithelial and stromal regions of the xenografts and original patient tumour. Results: IFP and nodal disease were not correlated with tumor engraftment. IFP measurements in the xenografts were generally lower than those in the patient’s tumor. Lymphatic metastasis increased with passage number as did levels of hypoxia in the epithelial component of the xenografts. The blood vessel density in the stromal component of the xenografts increased in parallel. When all the markers were compared between the biopsy and the respective 3rd generation xenograft 10 of 11 tumors showed a good correlation. Conclusions: This ongoing study provides characterization about tumoral and stromal heterogeneity in a unique orthotopic xenograft model.
Collapse
|
31
|
Zhou L, El-Deiry W, Wang W, Ingram ME, Katz SI. Extracellular protease imaging for cell mass tracking of xenografted human malignant pleural mesothelioma. Oncol Rep 2012; 28:883-8. [PMID: 22751990 DOI: 10.3892/or.2012.1888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 02/20/2012] [Indexed: 11/06/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is locally aggressive and challenging to quantitate non-invasively in vivo, particularly in orthotopic models of disease. We describe imaging of extracellular protease activity, typically elevated in locally aggressive tumors, as a novel method for tracking MPM in vivo. Mice bearing human MPM subcutaneous flank xenografted tumors were imaged with ProSense 680, an optical imaging agent of extracellular cysteine protease activity. The relative contribution of extracellular cysteine proteases to the ProSense tumor signal was estimated using RT-PCR quantitation of cysteine protease RNA expression of the MPM cell lines and compared to ArrayExpress microarray RNA expression data from human MPM tumors. Feasibility of orthotopic intraperitoneal MPM cell mass tracking with fluorescence signal was evaluated using CellVue Maroon-coated MSTO-211H and compared to bioluminescent signal using luciferase-transfected MSTO-211H cells. ProSense 680 yielded a robust tumor signal in MPM subcutaneous grafts, primarily resulting from MPM secretion of cathepsin L demonstrated not only by RT-PCR data on MPM cell lines but also by microarray expression data from resected human patient tumors. CellVue Maroon intraperitoneal tumor signal was robust and durable indicating feasibility of intraperitoneal cell mass tracking of orthotopically-xenografted MPM. Optical imaging of extracellular cysteine protease activity is useful for tracking MPM tumor cell mass in vivo. Intraperitoneal MPM cell mass tracking of fluorescently labeled tumor is feasible.
Collapse
Affiliation(s)
- Lanlan Zhou
- Department of Medicine (Hematology/Oncology), Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Penn State College of Medicine, Penn State Hershey Cancer Institute, Penn State Hershey Medical Center, Hershey, PA, USA
| | | | | | | | | |
Collapse
|
32
|
Abstract
Progress in oncology drug development has been hampered by a lack of preclinical models that reliably predict clinical activity of novel compounds in cancer patients. In an effort to address these shortcomings, there has been a recent increase in the use of patient-derived tumour xenografts (PDTX) engrafted into immune-compromised rodents such as athymic nude or NOD/SCID mice for preclinical modelling. Numerous tumour-specific PDTX models have been established and, importantly, they are biologically stable when passaged in mice in terms of global gene-expression patterns, mutational status, metastatic potential, drug responsiveness and tumour architecture. These characteristics might provide significant improvements over standard cell-line xenograft models. This Review will discuss specific PDTX disease examples illustrating an overview of the opportunities and limitations of these models in cancer drug development, and describe concepts regarding predictive biomarker development and future applications.
Collapse
|