1
|
Shi W, Tanzhu G, Chen L, Ning J, Wang H, Xiao G, Peng H, Jing D, Liang H, Nie J, Yi M, Zhou R. Radiotherapy in Preclinical Models of Brain Metastases: A Review and Recommendations for Future Studies. Int J Biol Sci 2024; 20:765-783. [PMID: 38169621 PMCID: PMC10758094 DOI: 10.7150/ijbs.91295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
Brain metastases (BMs) frequently occur in primary tumors such as lung cancer, breast cancer, and melanoma, and are associated with notably short natural survival. In addition to surgical interventions, chemotherapy, targeted therapy, and immunotherapy, radiotherapy (RT) is a crucial treatment for BM and encompasses whole-brain radiotherapy (WBRT) and stereotactic radiosurgery (SRS). Validating the efficacy and safety of treatment regimens through preclinical models is imperative for successful translation to clinical application. This not only advances fundamental research but also forms the theoretical foundation for clinical study. This review, grounded in animal models of brain metastases (AM-BM), explores the theoretical underpinnings and practical applications of radiotherapy in combination with chemotherapy, targeted therapy, immunotherapy, and emerging technologies such as nanomaterials and oxygen-containing microbubbles. Initially, we provided a concise overview of the establishment of AM-BMs. Subsequently, we summarize key RT parameters (RT mode, dose, fraction, dose rate) and their corresponding effects in AM-BMs. Finally, we present a comprehensive analysis of the current research status and future directions for combination therapy based on RT. In summary, there is presently no standardized regimen for AM-BM treatment involving RT. Further research is essential to deepen our understanding of the relationships between various parameters and their respective effects.
Collapse
Affiliation(s)
- Wen Shi
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Guilong Tanzhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Liu Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Jiaoyang Ning
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Hongji Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Gang Xiao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Haiqin Peng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Di Jing
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Huadong Liang
- Department of Technology, Hunan SJA Laboratory Animal Co., Ltd., Changsha, Hunan Province, China
| | - Jing Nie
- Department of Technology, Hunan SJA Laboratory Animal Co., Ltd., Changsha, Hunan Province, China
| | - Min Yi
- Department of Technology, Hunan SJA Laboratory Animal Co., Ltd., Changsha, Hunan Province, China
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| |
Collapse
|
2
|
Slika H, Karimov Z, Alimonti P, Abou-Mrad T, De Fazio E, Alomari S, Tyler B. Preclinical Models and Technologies in Glioblastoma Research: Evolution, Current State, and Future Avenues. Int J Mol Sci 2023; 24:16316. [PMID: 38003507 PMCID: PMC10671665 DOI: 10.3390/ijms242216316] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Glioblastoma is the most common malignant primary central nervous system tumor and one of the most debilitating cancers. The prognosis of patients with glioblastoma remains poor, and the management of this tumor, both in its primary and recurrent forms, remains suboptimal. Despite the tremendous efforts that are being put forward by the research community to discover novel efficacious therapeutic agents and modalities, no major paradigm shifts have been established in the field in the last decade. However, this does not mirror the abundance of relevant findings and discoveries made in preclinical glioblastoma research. Hence, developing and utilizing appropriate preclinical models that faithfully recapitulate the characteristics and behavior of human glioblastoma is of utmost importance. Herein, we offer a holistic picture of the evolution of preclinical models of glioblastoma. We further elaborate on the commonly used in vitro and vivo models, delving into their development, favorable characteristics, shortcomings, and areas of potential improvement, which aids researchers in designing future experiments and utilizing the most suitable models. Additionally, this review explores progress in the fields of humanized and immunotolerant mouse models, genetically engineered animal models, 3D in vitro models, and microfluidics and highlights promising avenues for the future of preclinical glioblastoma research.
Collapse
Affiliation(s)
- Hasan Slika
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
| | - Ziya Karimov
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
- Faculty of Medicine, Ege University, 35100 Izmir, Turkey
| | - Paolo Alimonti
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy; (P.A.); (E.D.F.)
| | - Tatiana Abou-Mrad
- Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon;
- Department of Neurosurgery, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Emerson De Fazio
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy; (P.A.); (E.D.F.)
| | - Safwan Alomari
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
| |
Collapse
|
3
|
Molecular Characterization of the Dual Effect of the GPER Agonist G-1 in Glioblastoma. Int J Mol Sci 2022; 23:ijms232214309. [PMID: 36430793 PMCID: PMC9695951 DOI: 10.3390/ijms232214309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary brain tumor in adults. Despite conventional treatment, consisting of a chirurgical resection followed by concomitant radio-chemotherapy, the 5-year survival rate is less than 5%. Few risk factors are clearly identified, but women are 1.4-fold less affected than men, suggesting that hormone and particularly estrogen signaling could have protective properties. Indeed, a high GPER1 (G-protein-coupled estrogen receptor) expression is associated with better survival, especially in women who produce a greater amount of estrogen. Therefore, we addressed the anti-tumor effect of the GPER agonist G-1 in vivo and characterized its molecular mechanism of action in vitro. First, the antiproliferative effect of G-1 was confirmed in a model of xenografted nude mice. A transcriptome analysis of GBM cells exposed to G-1 was performed, followed by functional analysis of the differentially expressed genes. Lipid and steroid synthesis pathways as well as cell division processes were both affected by G-1, depending on the dose and duration of the treatment. ANGPTL4, the first marker of G-1 exposure in GBM, was identified and validated in primary GBM cells and patient samples. These data strongly support the potential of G-1 as a promising chemotherapeutic compound for the treatment of GBM.
Collapse
|
4
|
Xiang J, Ma L, Gu Z, Jin H, Zhai H, Tong J, Liang T, Li J, Ren Q, Liu Q. A Boronated Derivative of Temozolomide Showing Enhanced Efficacy in Boron Neutron Capture Therapy of Glioblastoma. Cells 2022; 11:cells11071173. [PMID: 35406737 PMCID: PMC8998031 DOI: 10.3390/cells11071173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 02/04/2023] Open
Abstract
There is an incontestable need for improved treatment modality for glioblastoma due to its extraordinary resistance to traditional chemoradiation therapy. Boron neutron capture therapy (BNCT) may play a role in the future. We designed and synthesized a 10B-boronated derivative of temozolomide, TMZB. BNCT was carried out with a total neutron radiation fluence of 2.4 ± 0.3 × 1011 n/cm2. The effects of TMZB in BNCT were measured with a clonogenic cell survival assay in vitro and PET/CT imaging in vivo. Then, 10B-boronated phenylalanine (BPA) was tested in parallel with TMZB for comparison. The IC50 of TMZB for the cytotoxicity of clonogenic cells in HS683 was 0.208 mM, which is comparable to the IC50 of temozolomide at 0.213 mM. In BNCT treatment, 0.243 mM TMZB caused 91.2% ± 6.4% of clonogenic cell death, while 0.239 mM BPA eliminated 63.7% ± 6.3% of clonogenic cells. TMZB had a tumor-to-normal brain ratio of 2.9 ± 1.1 and a tumor-to-blood ratio of 3.8 ± 0.2 in a mouse glioblastoma model. BNCT with TMZB in this model caused 58.2% tumor shrinkage at 31 days after neutron irradiation, while the number for BPA was 35.2%. Therefore, by combining the effects of chemotherapy from temozolomide and radiotherapy with heavy charged particles from BNCT, TMZB-based BNCT exhibited promising potential for therapeutic applications in glioblastoma treatment.
Collapse
Affiliation(s)
- Jing Xiang
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China;
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen 518132, China;
| | - Lin Ma
- Department of Stomatology, General Hospital, Shenzhen University, Shenzhen 518055, China;
| | - Zheng Gu
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen 518132, China;
| | - Hongjun Jin
- Guangdong Provincial Key Lab of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China;
| | - Hongbin Zhai
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China;
| | - Jianfei Tong
- Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (J.T.); (T.L.); (J.L.)
- Spallation Neutron Source Science Center, Dongguan 523803, China
| | - Tianjiao Liang
- Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (J.T.); (T.L.); (J.L.)
- Spallation Neutron Source Science Center, Dongguan 523803, China
| | - Juan Li
- Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (J.T.); (T.L.); (J.L.)
- Spallation Neutron Source Science Center, Dongguan 523803, China
| | - Qiushi Ren
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China;
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen 518132, China;
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China;
- Correspondence: (Q.R.); (Q.L.); Tel.: +86-0755-26038837 (Q.R. & Q.L.)
| | - Qi Liu
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen 518132, China;
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China;
- Correspondence: (Q.R.); (Q.L.); Tel.: +86-0755-26038837 (Q.R. & Q.L.)
| |
Collapse
|
5
|
Kim JH, Lee E, Yun J, Ryu HS, Kim HK, Ju YW, Kim K, Kim J, Moon H. Calsequestrin 2 overexpression in breast cancer increases tumorigenesis and metastasis by modulating the tumor microenvironment. Mol Oncol 2022; 16:466-484. [PMID: 34743414 PMCID: PMC8763655 DOI: 10.1002/1878-0261.13136] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 10/05/2021] [Accepted: 11/04/2021] [Indexed: 01/16/2023] Open
Abstract
The spatial tumor shape is determined by the complex interactions between tumor cells and their microenvironment. Here, we investigated the role of a newly identified breast cancer-related gene, calsequestrin 2 (CASQ2), in tumor-microenvironment interactions during tumor growth and metastasis. We analyzed gene expression and three-dimensional tumor shape data from the breast cancer dataset of The Cancer Genome Atlas (TCGA) and identified CASQ2 as a potential regulator of tumor-microenvironment interaction. In TCGA breast cancer cases containing information of three-dimensional tumor shapes, CASQ2 mRNA showed the highest correlation with the spatial tumor shapes. Furthermore, we investigated the expression pattern of CASQ2 in human breast cancer tissues. CASQ2 was not detected in breast cancer cell lines in vitro but was induced in the xenograft tumors and human breast cancer tissues. To evaluate the role of CASQ2, we established CASQ2-overexpressing breast cancer cell lines for in vitro and in vivo experiments. CASQ2 overexpression in breast cancer cells resulted in a more aggressive phenotype and altered epithelial-mesenchymal transition (EMT) markers in vitro. CASQ2 overexpression induced cancer-associated fibroblast characteristics along with increased hypoxia-inducible factor 1α (HIF1α) expression in stromal fibroblasts. CASQ2 overexpression accelerated tumorigenesis, induced collagen structure remodeling, and increased distant metastasis in vivo. CASQ2 conferred more metaplastic features to triple-negative breast cancer cells. Our data suggest that CASQ2 is a key regulator of breast cancer tumorigenesis and metastasis by modulating diverse aspects of tumor-microenvironment interactions.
Collapse
Affiliation(s)
- Ju Hee Kim
- Biomedical Research InstituteSeoul National University HospitalSouth Korea
| | - Eun‐Shin Lee
- Biomedical Research InstituteSeoul National University HospitalSouth Korea
- Department of PathologySeoul National University School of MedicineSouth Korea
| | - Jihui Yun
- Genomic Medicine InstituteMedical Research CenterSeoul National UniversityKorea
- Department of Biomedical SciencesSeoul National University College of MedicineKorea
| | - Han Suk Ryu
- Department of PathologySeoul National University HospitalSouth Korea
| | - Hong Kyu Kim
- Department of SurgerySeoul National University HospitalKorea
| | - Young Wook Ju
- Department of SurgerySeoul National University HospitalKorea
| | - Kwangsoo Kim
- Division of Clinical BioinformaticsSeoul National University HospitalKorea
| | - Jong‐Il Kim
- Genomic Medicine InstituteMedical Research CenterSeoul National UniversityKorea
- Department of Biomedical SciencesSeoul National University College of MedicineKorea
- Cancer Research InstituteSeoul National UniversityKorea
- Department of Biochemistry and Molecular BiologySeoul National University College of MedicineKorea
| | - Hyeong‐Gon Moon
- Department of SurgerySeoul National University HospitalKorea
- Cancer Research InstituteSeoul National UniversityKorea
- Department of SurgerySeoul National University College of MedicineSouth Korea
| |
Collapse
|
6
|
Preclinical models of glioblastoma: limitations of current models and the promise of new developments. Expert Rev Mol Med 2021; 23:e20. [PMID: 34852856 DOI: 10.1017/erm.2021.20] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumour, yet little progress has been made towards providing better treatment options for patients diagnosed with this devastating condition over the last few decades. The complex nature of the disease, heterogeneity, highly invasive potential of GBM tumours and until recently, reduced investment in research funding compared with other cancer types, are contributing factors to few advancements in disease management. Survival rates remain low with less than 5% of patients surviving 5 years. Another important contributing factor is the use of preclinical models that fail to fully recapitulate GBM pathophysiology, preventing efficient translation from the lab into successful therapies in the clinic. This review critically evaluates current preclinical GBM models, highlighting advantages and disadvantages of using such models, and outlines several emerging techniques in GBM modelling using animal-free approaches. These novel approaches to a highly complex disease such as GBM show evidence of a more truthful recapitulation of GBM pathobiology with high reproducibility. The resulting advancements in this field will offer new biological insights into GBM and its aetiology with potential to contribute towards the development of much needed improved treatments for GBM in future.
Collapse
|
7
|
Haddad AF, Young JS, Amara D, Berger MS, Raleigh DR, Aghi MK, Butowski NA. Mouse models of glioblastoma for the evaluation of novel therapeutic strategies. Neurooncol Adv 2021; 3:vdab100. [PMID: 34466804 PMCID: PMC8403483 DOI: 10.1093/noajnl/vdab100] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Glioblastoma (GBM) is an incurable brain tumor with a median survival of approximately 15 months despite an aggressive standard of care that includes surgery, chemotherapy, and ionizing radiation. Mouse models have advanced our understanding of GBM biology and the development of novel therapeutic strategies for GBM patients. However, model selection is crucial when testing developmental therapeutics, and each mouse model of GBM has unique advantages and disadvantages that can influence the validity and translatability of experimental results. To shed light on this process, we discuss the strengths and limitations of 3 types of mouse GBM models in this review: syngeneic models, genetically engineered mouse models, and xenograft models, including traditional xenograft cell lines and patient-derived xenograft models.
Collapse
Affiliation(s)
- Alexander F Haddad
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Jacob S Young
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Dominic Amara
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - David R Raleigh
- Department of Neurological Surgery, University of California, San Francisco, California, USA
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California, USA
| | - Manish K Aghi
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Nicholas A Butowski
- Department of Neurological Surgery, University of California, San Francisco, California, USA
- Corresponding Author: Nicholas A. Butowski, MD, Department of Neurological Surgery, University of California, San Francisco, 400 Parnassus Ave Eighth Floor, San Francisco, CA, 94143, USA ()
| |
Collapse
|
8
|
Serafim RB, da Silva P, Cardoso C, Di Cristofaro LFM, Netto RP, de Almeida R, Navegante G, Storti CB, de Sousa JF, de Souza FC, Panepucci R, Moreira CG, Penna LS, Silva WA, Valente V. Expression Profiling of Glioblastoma Cell Lines Reveals Novel Extracellular Matrix-Receptor Genes Correlated With the Responsiveness of Glioma Patients to Ionizing Radiation. Front Oncol 2021; 11:668090. [PMID: 34211843 PMCID: PMC8240593 DOI: 10.3389/fonc.2021.668090] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/30/2021] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is the most lethal and frequent type of brain tumor, leading patients to death in approximately 14 months after diagnosis. GBM treatment consists in surgical removal followed by radio and chemotherapy. However, tumors commonly relapse and the treatment promotes only a slight increase in patient survival. Thus, uncovering the cellular mechanisms involved in GBM resistance is of utmost interest, and the use of cell lines has been shown to be an extremely important tool. In this work, the exploration of RNAseq data from different GBM cell lines revealed different expression signatures, distinctly correlated with the behavior of GBM cell lines regarding proliferation indexes and radio-resistance. U87MG and U138MG cells, which presented expressively reduced proliferation and increased radio-resistance, showed a particular expression signature encompassing enrichment in many extracellular matrix (ECM) and receptor genes. Contrasting, U251MG and T98G cells, that presented higher proliferation and sensibility to radiation, exhibited distinct signatures revealing consistent enrichments for DNA repair processes and although several genes from the ECM-receptor pathway showed up-regulation, enrichments for this pathway were not detected. The ECM-receptor is a master regulatory pathway that is known to impact several cellular processes including: survival, proliferation, migration, invasion, and DNA damage signaling and repair, corroborating the associations we found. Furthermore, searches to The Cancer Genome Atlas (TCGA) repository revealed prognostic correlations with glioma patients for the majority of genes highlighted in the signatures and led to the identification of 31 ECM-receptor genes individually correlated with radiation responsiveness. Interestingly, we observed an association between the number of upregulated genes and survivability greater than 5 years after diagnosis, where almost all the patients that presented 21 or more upregulated genes were deceased before 5 years. Altogether our findings suggest the clinical relevance of ECM-receptor genes signature found here for radiotherapy decision and as biomarkers of glioma prognosis.
Collapse
Affiliation(s)
- Rodolfo Bortolozo Serafim
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
- Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Patrick da Silva
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Cibele Cardoso
- Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | | | - Renato Petitto Netto
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Rodrigo de Almeida
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Geovana Navegante
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Camila Baldin Storti
- Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Juliana Ferreira de Sousa
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Felipe Canto de Souza
- Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Rodrigo Panepucci
- Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | | | - Larissa Siqueira Penna
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Wilson Araujo Silva
- Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Valeria Valente
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
- Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| |
Collapse
|
9
|
Kwon ATJ, Mohri K, Takizawa S, Arakawa T, Takahashi M, Kaczkowski B, Furuno M, Suzuki H, Tagami S, Mukai H, Arner E. Development of p53 knockout U87MG cell line for unbiased drug delivery testing system using CRISPR-Cas9 and transcriptomic analysis. J Biotechnol 2021; 332:72-82. [PMID: 33836165 DOI: 10.1016/j.jbiotec.2021.03.017] [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: 10/06/2020] [Revised: 03/15/2021] [Accepted: 03/23/2021] [Indexed: 10/21/2022]
Abstract
Antibody-drug conjugates offers many advantages as a drug delivery platform that allows for highly specific targeting of cell types and genes. Ideally, testing the efficacy of these systems requires two cell types to be different only in the gene targeted by the drug, with the rest of the cellular machinery unchanged, in order to minimize other potential differences from obscuring the effects of the drug. In this study, we created multiple variants of U87MG cells with targeted mutation in the TP53 gene using the CRISPR-Cas9 system, and determined that their major transcriptional differences stem from the loss of p53 function. Using the transcriptome data, we predicted which mutant clones would have less divergent phenotypes from the wild type and thereby serve as the best candidates to be used as drug delivery testing platforms. Further in vitro and in vivo assays of cell morphology, proliferation rate and target antigen-mediated uptake supported our predictions. Based on the combined analysis results, we successfully selected the best qualifying mutant clone. This study serves as proof-of-principle of the approach and paves the way for extending to additional cell types and target genes.
Collapse
Affiliation(s)
| | - Kohta Mohri
- RIKEN Center for Biosystems Dynamic Research, Japan
| | | | | | | | | | | | | | | | | | - Erik Arner
- RIKEN Center for Integrative Medical Sciences, Japan.
| |
Collapse
|
10
|
Baschnagel AM, Kaushik S, Durmaz A, Goldstein S, Ong IM, Abel L, Clark PA, Gurel Z, Leal T, Buehler D, Iyer G, Scott JG, Kimple RJ. Development and characterization of patient-derived xenografts from non-small cell lung cancer brain metastases. Sci Rep 2021; 11:2520. [PMID: 33510214 PMCID: PMC7843608 DOI: 10.1038/s41598-021-81832-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/12/2021] [Indexed: 01/08/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) brain metastasis cell lines and in vivo models are not widely accessible. Herein we report on a direct-from patient-derived xenograft (PDX) model system of NSCLC brain metastases with genomic annotation useful for translational and mechanistic studies. Both heterotopic and orthotopic intracranial xenografts were established and RNA and DNA sequencing was performed on patient and matching tumors. Morphologically, strong retention of cytoarchitectural features was observed between original patient tumors and PDXs. Transcriptome and mutation analysis revealed high correlation between matched patient and PDX samples with more than more than 95% of variants detected being retained in the matched PDXs. PDXs demonstrated response to radiation, response to selumetinib in tumors harboring KRAS G12C mutations and response to savolitinib in a tumor with MET exon 14 skipping mutation. Savolitinib also demonstrated in vivo radiation enhancement in our MET exon 14 mutated PDX. Early passage cell strains showed high consistency between patient and PDX tumors. Together, these data describe a robust human xenograft model system for investigating NSCLC brain metastases. These PDXs and cell lines show strong phenotypic and molecular correlation with the original patient tumors and provide a valuable resource for testing preclinical therapeutics.
Collapse
Affiliation(s)
- Andrew M Baschnagel
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, K4/B100-0600, Madison, WI, 53792, USA.
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA.
| | - Saakshi Kaushik
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, K4/B100-0600, Madison, WI, 53792, USA
| | - Arda Durmaz
- Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Steve Goldstein
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Irene M Ong
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Lindsey Abel
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, K4/B100-0600, Madison, WI, 53792, USA
| | - Paul A Clark
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, K4/B100-0600, Madison, WI, 53792, USA
| | - Zafer Gurel
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, K4/B100-0600, Madison, WI, 53792, USA
| | - Ticiana Leal
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Division of Hematology/Oncology, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Darya Buehler
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Gopal Iyer
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, K4/B100-0600, Madison, WI, 53792, USA
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Jacob G Scott
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, 10201 Carnegie Ave, Cleveland, OH, 44195, USA
| | - Randall J Kimple
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, K4/B100-0600, Madison, WI, 53792, USA.
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA.
- , 3107 WIMR, 1111 Highland Ave., Madison, WI, 53705, USA.
| |
Collapse
|
11
|
Herbener VJ, Burster T, Goreth A, Pruss M, von Bandemer H, Baisch T, Fitzel R, Siegelin MD, Karpel-Massler G, Debatin KM, Westhoff MA, Strobel H. Considering the Experimental use of Temozolomide in Glioblastoma Research. Biomedicines 2020; 8:E151. [PMID: 32512726 PMCID: PMC7344626 DOI: 10.3390/biomedicines8060151] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/30/2020] [Accepted: 05/31/2020] [Indexed: 12/17/2022] Open
Abstract
Temozolomide (TMZ) currently remains the only chemotherapeutic component in the approved treatment scheme for Glioblastoma (GB), the most common primary brain tumour with a dismal patient's survival prognosis of only ~15 months. While frequently described as an alkylating agent that causes DNA damage and thus-ultimately-cell death, a recent debate has been initiated to re-evaluate the therapeutic role of TMZ in GB. Here, we discuss the experimental use of TMZ and highlight how it differs from its clinical role. Four areas could be identified in which the experimental data is particularly limited in its translational potential: 1. transferring clinical dosing and scheduling to an experimental system and vice versa; 2. the different use of (non-inert) solvent in clinic and laboratory; 3. the limitations of established GB cell lines which only poorly mimic GB tumours; and 4. the limitations of animal models lacking an immune response. Discussing these limitations in a broader biomedical context, we offer suggestions as to how to improve transferability of data. Finally, we highlight an underexplored function of TMZ in modulating the immune system, as an example of where the aforementioned limitations impede the progression of our knowledge.
Collapse
Affiliation(s)
- Verena J. Herbener
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Timo Burster
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan 010000, Kazakhstan;
| | - Alicia Goreth
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Maximilian Pruss
- Department of Gynecology and Obstetrics, Medical Faculty, University Hospital of the Heinrich-Heine-University Duesseldorf, D-40225 Duesseldorf, Germany;
- Department of Neurosurgery, University Medical Center Ulm, D-89081 Ulm, Germany;
| | - Hélène von Bandemer
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Tim Baisch
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Rahel Fitzel
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Markus D. Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA;
| | - Georg Karpel-Massler
- Department of Neurosurgery, University Medical Center Ulm, D-89081 Ulm, Germany;
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Hannah Strobel
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| |
Collapse
|
12
|
Olig2 SUMOylation protects against genotoxic damage response by antagonizing p53 gene targeting. Cell Death Differ 2020; 27:3146-3161. [PMID: 32483381 PMCID: PMC7560653 DOI: 10.1038/s41418-020-0569-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 01/05/2023] Open
Abstract
Posttranslational modifications of nuclear proteins, including transcription factors, nuclear receptors, and their coregulators, have attracted much attention in cancer research. Although phosphorylation of oligodendrocyte transcription factor 2 (Olig2) may contribute to the notorious resistance of gliomas to radiation and genotoxic drugs, the precise mechanisms remain elusive. We show here that in addition to phosphorylation, Olig2 is also conjugated by small ubiquitin-like modifier-1 (SUMO1) at three lysine residues K27, K76, and K112. SUMOylation is required for Olig2 to suppress p53-mediated cell cycle arrest and apoptosis induced by genotoxic damage, and to enhance resistance to temozolomide (TMZ) in glioma. Both SUMOylation and triple serine motif (TSM) phosphorylation of Olig2 are required for the antiapoptotic function. Olig2 SUMOylation enhances its genetic targeting ability, which in turn occludes p53 recruitment to Cdkn1a promoter for DNA-damage responses. Our work uncovers a SUMOylation-dependent regulatory mechanism of Olig2 in regulating cancer survival.
Collapse
|
13
|
Polyphyllin VII Promotes Apoptosis and Autophagic Cell Death via ROS-Inhibited AKT Activity, and Sensitizes Glioma Cells to Temozolomide. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1805635. [PMID: 31814867 PMCID: PMC6877958 DOI: 10.1155/2019/1805635] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/07/2019] [Accepted: 09/23/2019] [Indexed: 02/05/2023]
Abstract
The high recurrence frequency of gliomas but deficiency of effective treatment and prevalent chemoresistance have elicited interests in exploring and developing new agents. Paris polyphyllins are monomers extracted from rhizome of Paris polyphylla var. yunnanensis. Here, we first reported that polyphyllin VII (PP7) exhibited cytotoxic effect on glioma cells. PP7 significantly suppressed the viability and induced cell death of U87-MG and U251 cells after 24 h, with the IC50 values 4.24 ± 0.87 μM and 2.17 ± 0.14 μM, respectively. Both apoptotic and autophagic processes were involved in the cytotoxic effect of PP7, as PP7 activated the Bcl2/Bax pathway and the inhibition of autophagy partly rescued the toxicity of PP7 in glioma cells. In addition, an inhibition of AKT/mTORC1 activity was found after PP7 administration, and it seemed that the overproduction of reactive oxygen species (ROS) was responsible for this effect. Namely, the removal of ROS by NAC treatment mitigated PP7-induced cell death, autophagy, and its effect on the AKT/mTORC1 signaling. Additionally, a combination assay of PP7 with temozolomide (TMZ), the most used chemotherapy for glioma patients, was performed resulting in synergism, while PP7 reduced TMZ resistance through inhibition of MGMT expression. Thus, our study reports PP7 as a potential agent for glioma treatment and reveals its underlying mechanisms of action.
Collapse
|
14
|
Rastgar S, Alijani Ardeshir R, Zabihi E, Movahedinia A, Salati AP. Immunotoxicity of estrogen and nonylphenol on apoptosis and expression of ERs in goldfish macrophage: Opening new avenue for discovering the role of experimental model systems and sexes. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 209:159-167. [PMID: 30780113 DOI: 10.1016/j.aquatox.2019.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/08/2019] [Indexed: 06/09/2023]
Abstract
The expression of estrogen receptors (ERs) and their roles in important cell processes such as apoptosis in the macrophages exposed to estrogen/xenoestrogen have remained a complex secret. This study focused on the expression of estrogen receptors (ERs) and the stimulation of apoptosis in the macrophages from the two sexes of goldfish (Carassius auratus) exposed to 17-βestradiol (E2) and nonylphenol (NP) under in vivo and in vitro conditions. For the in vivo experiment, fish were exposed to NP (10-6 M and 10-7 M) and E2 (10-6 M) for 24 days. Then, the head kidney macrophages from the male and the female goldfish were isolated and assayed. For the in vitro experiments, the macrophages derived from the two sexes were cultured in L-15 medium and exposed to E2 (150 nM) and NP (10 nM and 150 nM) for 3 days. The results showed that the three isoforms of ERs (ERα, ERβ1, ERβ2) were expressed in the goldfish macrophages. After the exposure of macrophages to NP and E2, sex-specific increase of ERs expression and apoptosis were observed (P < 0.05). The expression of ERα after NP treatment showed the highest alteration, with the response being concentration-dependent. The most alteration of ERs expression were observed in the in vivo experiment. This study provides insight to understand how exposure of the goldfish macrophages to E2 and NP can up-regulate the transcript levels of estrogen receptor subtypes and stimulate apoptosis.
Collapse
Affiliation(s)
- Sara Rastgar
- Department of Marine Biology, Faculty of Marine Sciences, Khorramshahr University of Marine Science and Technology, P.O. Box 669, Khorramshahr, Iran
| | - Rashid Alijani Ardeshir
- Department of Marine Biology, Faculty of Marine Sciences, Khorramshahr University of Marine Science and Technology, P.O. Box 669, Khorramshahr, Iran
| | - Ebrahim Zabihi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abdolali Movahedinia
- Department of Marine Biology, Faculty of Marine Sciences, University of Mazandaran, Babolsar, Iran
| | - Amir Parviz Salati
- Department of Fisheries, Faculty of Marine Natural resources, Khorramshahr University of Marine Science and Technology, P.O. Box 669, Khorramshahr, Iran.
| |
Collapse
|
15
|
Evolution of the Supermodel: Progress in Modelling Radiotherapy Response in Mice. Clin Oncol (R Coll Radiol) 2019; 31:272-282. [PMID: 30871751 DOI: 10.1016/j.clon.2019.02.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 02/14/2019] [Accepted: 02/15/2019] [Indexed: 12/18/2022]
Abstract
Mouse models are essential tools in cancer research that have been used to understand the genetic basis of tumorigenesis, cancer progression and to test the efficacies of anticancer treatments including radiotherapy. They have played a critical role in our understanding of radiotherapy response in tumours and normal tissues and continue to evolve to better recapitulate the underlying biology of humans. In addition, recent developments in small animal irradiators have significantly improved in vivo irradiation techniques, allowing previously unimaginable experimental approaches to be explored in the laboratory. The combination of contemporary mouse models with small animal irradiators represents a major step forward for the radiobiology field in being able to much more accurately replicate clinical exposure scenarios. As radiobiology studies become ever more sophisticated in reflecting developments in the clinic, it is increasingly important to understand the basis and potential limitations of extrapolating data from mice to humans. This review provides an overview of mouse models and small animal radiotherapy platforms currently being used as advanced radiobiological research tools towards improving the translational power of preclinical studies.
Collapse
|
16
|
Towner RA, Smith N, Saunders D, Brown CA, Cai X, Ziegler J, Mallory S, Dozmorov MG, Coutinho De Souza P, Wiley G, Kim K, Kang S, Kong DS, Kim YT, Fung KM, Wren JD, Battiste J. OKN-007 Increases temozolomide (TMZ) Sensitivity and Suppresses TMZ-Resistant Glioblastoma (GBM) Tumor Growth. Transl Oncol 2019; 12:320-335. [PMID: 30468988 PMCID: PMC6251232 DOI: 10.1016/j.tranon.2018.10.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/28/2018] [Accepted: 10/01/2018] [Indexed: 02/06/2023] Open
Abstract
Treatment of glioblastoma (GBM) remains a challenge using conventional chemotherapy, such as temozolomide (TMZ), and is often ineffective as a result of drug resistance. We have assessed a novel nitrone-based agent, OKN-007, and found it to be effective in decreasing tumor volumes and increasing survival in orthotopic GBM xenografts by decreasing cell proliferation and angiogenesis and increasing apoptosis. In this study, we assessed combining OKN-007 with TMZ in vivo in a human G55 GBM orthotopic xenograft model and in vitro in TMZ-resistant and TMZ-sensitive human GBM cell lines. For the in vivo studies, magnetic resonance imaging was used to assess tumor growth and vascular alterations. Percent animal survival was also determined. For the in vitro studies, cell growth, IC50 values, RNA-seq, RT-PCR, and ELISA were used to assess growth inhibition, possible mechanism-of actions (MOAs) associated with combined OKN-007 + TMZ versus TMZ alone, and gene and protein expression levels, respectively. Microarray analysis of OKN-007-treated rat F98 glioma tumors was also carried out to determine possible MOAs of OKN-007 in glioma-bearing animals either treated or not treated with OKN-007. OKN-007 seems to elicit its effect on GBM tumors via inhibition of tumorigenic TGF-β1, which affects the extracellular matrix. When combined with TMZ, OKN-007 significantly increases percent survival, decreases tumor volumes, and normalizes tumor blood vasculature in vivo compared to untreated tumors and seems to affect TMZ-resistant GBM cells possibly via IDO-1, SUMO2, and PFN1 in vitro. Combined OKN-007 + TMZ may be a potentially potent treatment strategy for GBM patients.
Collapse
Affiliation(s)
- Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of PathologyUniversity of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Chase A Brown
- Arthritis and Clinical Immunology Research Program, Division of Genomics and Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Xue Cai
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jadith Ziegler
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of PathologyUniversity of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Mikhail G Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Graham Wiley
- Clinical Genomics Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kyeongsoon Kim
- Department of Pharmaceutical Engineering, Inje University, Gimhae-si, Gyeongsangnam-do, Republic of Korea; Oblato, Inc., Princeton, NJ, USA
| | | | - Doo-Sik Kong
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Young-Tae Kim
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, USA
| | - Kar-Ming Fung
- Department of PathologyUniversity of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jonathan D Wren
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Arthritis and Clinical Immunology Research Program, Division of Genomics and Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - James Battiste
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of NeurologyUniversity of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
17
|
Gargiulo G. Next-Generation in vivo Modeling of Human Cancers. Front Oncol 2018; 8:429. [PMID: 30364119 PMCID: PMC6192385 DOI: 10.3389/fonc.2018.00429] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/13/2018] [Indexed: 12/19/2022] Open
Abstract
Animal models of human cancers played a major role in our current understanding of tumor biology. In pre-clinical oncology, animal models empowered drug target and biomarker discovery and validation. In turn, this resulted in improved care for cancer patients. In the quest for understanding and treating a diverse spectrum of cancer types, technological breakthroughs in genetic engineering and single cell "omics" offer tremendous potential to enhance the informative value of pre-clinical models. Here, I review the state-of-the-art in modeling human cancers with focus on animal models for human malignant gliomas. The review highlights the use of glioma models in dissecting mechanisms of tumor initiation, in the retrospective identification of tumor cell-of-origin, in understanding tumor heterogeneity and in testing the potential of immuno-oncology. I build on the deep review of glioma models as a basis for a more general discussion of the potential ways in which transformative technologies may shape the next-generation of pre-clinical models. I argue that refining animal models along the proposed lines will benefit the success rate of translation for pre-clinical research in oncology.
Collapse
Affiliation(s)
- Gaetano Gargiulo
- Molecular Oncology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
18
|
El Alaoui-Lasmaili K, Faivre B. Antiangiogenic therapy: Markers of response, "normalization" and resistance. Crit Rev Oncol Hematol 2018; 128:118-129. [PMID: 29958627 DOI: 10.1016/j.critrevonc.2018.06.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 02/08/2023] Open
Abstract
Currently in cancer treatment, one premise is to use antiangiogenic therapies in association with chemotherapy or radiotherapy to augment their efficacy by benefiting from the vascular "normalization" induced by antiangiogenic therapy. This concept defines the time during which the tumor blood vessels adopt normal-like morphology and functionality, i.e. the blood vessels become more mature, the perfusion augments and hypoxia decreases. To date, there is such a diversity of treatment protocols where the type of antiangiogenic to adopt, its dose and duration of administration are different, that knowing when and how to treat is problematic. In this review, we analyzed thoroughly preclinical and clinical studies that use antiangiogenic treatments to benefit from the "normalization" and showed that the effects depend on the type of antiangiogenic administrated (anti-VEGF, anti-VEGFR, Multi-Kinase Inhibitor) and on the duration of treatment. Finally, biomarkers of "normalization" and resistance that could be used in the clinic are presented.
Collapse
Affiliation(s)
| | - Béatrice Faivre
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; Université de Lorraine, Faculté de Pharmacie, Nancy, France.
| |
Collapse
|
19
|
Quezada C, Torres Á, Niechi I, Uribe D, Contreras-Duarte S, Toledo F, San Martín R, Gutiérrez J, Sobrevia L. Role of extracellular vesicles in glioma progression. Mol Aspects Med 2018; 60:38-51. [PMID: 29222067 DOI: 10.1016/j.mam.2017.12.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/01/2017] [Accepted: 12/04/2017] [Indexed: 12/15/2022]
Abstract
The role of extracellular vesicles in cancer biology has emerged as a focus of the study of great importance and has been shown to directly influence tumour development in several cancers including brain tumours, such as gliomas. Gliomas are the most aggressive brain tumours, and in the last time, a considerable effort has been made to understand their biology. Studies focus in the signalling pathways involved in the processes of angiogenesis, viability, drug resistance and immune response evasion, as well as gliomas ability to infiltrate healthy tissue, a phenomenon regulated by the migratory and invasive capacity of the cells within a tumour. In this review, we summarize the different types and classifications of extracellular vesicles, their intravesicular content, and their role in the regulation of tumour progression processes in glioma.
Collapse
Affiliation(s)
- Claudia Quezada
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile.
| | - Ángelo Torres
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Ignacio Niechi
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Daniel Uribe
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Susana Contreras-Duarte
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Fernando Toledo
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán 3780000, Chile
| | - Rody San Martín
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Jaime Gutiérrez
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Cellular Signaling and Differentiation Laboratory (CSDL), School of Medical Technology, Health Sciences Faculty, Universidad San Sebastián, Santiago 7510157, Chile.
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029, Queensland, Australia.
| |
Collapse
|
20
|
Remšík J, Fedr R, Navrátil J, Binó L, Slabáková E, Fabian P, Svoboda M, Souček K. Plasticity and intratumoural heterogeneity of cell surface antigen expression in breast cancer. Br J Cancer 2018; 118:813-819. [PMID: 29462126 PMCID: PMC5886127 DOI: 10.1038/bjc.2017.497] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/19/2017] [Accepted: 12/20/2017] [Indexed: 12/20/2022] Open
Abstract
Background: The intratumoural heterogeneity, often driven by epithelial-to-mesenchymal transition (EMT), significantly contributes to chemoresistance and disease progression in adenocarcinomas. Methods: We introduced a high-throughput screening platform to identify surface antigens that associate with epithelial–mesenchymal plasticity in well-defined pairs of epithelial cell lines and their mesenchymal counterparts. Using multicolour flow cytometry, we then analysed the expression of 10 most robustly changed antigens and identified a 10-molecule surface signature, in pan-cytokeratin-positive/EpCAM-positive and -negative fractions of dissociated breast tumours. Results: We found that surface CD9, CD29, CD49c, and integrin β5 are lost in breast cancer cells that underwent EMT in vivo. The tetraspanin family member CD9 was concordantly downregulated both in vitro and in vivo and associated with epithelial phenotype and favourable prognosis. Conclusions: We propose that overall landscape of 10-molecule surface signature expression reflects the epithelial–mesenchymal plasticity in breast cancer.
Collapse
Affiliation(s)
- Ján Remšík
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Pekařská 53, Brno 656 91, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Radek Fedr
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Pekařská 53, Brno 656 91, Czech Republic
| | - Jiří Navrátil
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Žlutý kopec 7, Brno 656 53, Czech Republic
| | - Lucia Binó
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic
| | - Eva Slabáková
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic
| | - Pavel Fabian
- Department of Oncological Pathology, Masaryk Memorial Cancer Institute, Žlutý kopec 7, Brno 656 53, Czech Republic
| | - Marek Svoboda
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Žlutý kopec 7, Brno 656 53, Czech Republic
| | - Karel Souček
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Pekařská 53, Brno 656 91, Czech Republic
| |
Collapse
|
21
|
A Novel Cell Line Based Orthotopic Xenograft Mouse Model That Recapitulates Human Hepatoblastoma. Sci Rep 2017; 7:17751. [PMID: 29259231 PMCID: PMC5736579 DOI: 10.1038/s41598-017-17665-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 11/29/2017] [Indexed: 12/19/2022] Open
Abstract
Currently, preclinical testing of therapies for hepatoblastoma (HB) is limited to subcutaneous and intrasplenic xenograft models that do not recapitulate the hepatic tumors seen in patients. We hypothesized that injection of HB cell lines into the livers of mice would result in liver tumors that resemble their clinical counterparts. HepG2 and Huh-6 HB cell lines were injected, and tumor growth was monitored with bioluminescence imaging (BLI) and magnetic resonance imaging (MRI). Levels of human α-fetoprotein (AFP) were monitored in the serum of animals. Immunohistochemical and gene expression analyses were also completed on xenograft tumor samples. BLI signal indicative of tumor growth was seen in 55% of HepG2- and Huh-6-injected animals after a period of four to seven weeks. Increased AFP levels correlated with tumor growth. MRI showed large intrahepatic tumors with active neovascularization. HepG2 and Huh-6 xenografts showed expression of β-catenin, AFP, and Glypican-3 (GPC3). HepG2 samples displayed a consistent gene expression profile most similar to human HB tumors. Intrahepatic injection of HB cell lines leads to liver tumors in mice with growth patterns and biologic, histologic, and genetic features similar to human HB tumors. This orthotopic xenograft mouse model will enable clinically relevant testing of novel agents for HB.
Collapse
|
22
|
Wang F, Jiang H, Wang S, Chen B. Dual Functional MicroRNA-186-5p Targets both FGF2 and RelA to Suppress Tumorigenesis of Glioblastoma Multiforme. Cell Mol Neurobiol 2017; 37:1433-1442. [PMID: 28213656 PMCID: PMC11482140 DOI: 10.1007/s10571-017-0474-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/12/2017] [Indexed: 01/08/2023]
Abstract
Glioblastoma multiforme (GBM) is one of the most malignant cancers. MicroRNAs (miRs) were reported to play important roles in GBM recently. However, the role of a novel miR-186-5p in GBM tumorigenesis is still elusive. Using bioinformatics, miR-186-5p was identified as potential regulators of both fibroblast growth factor (FGF)-2 and NF-κB subunit RelA. Luciferase reporter assay was used to confirm the direct recognition FGF2 and RelA mRNAs by miR-186-5p. Invasion and migration assays were employed to study the effect of miR-186-5p on GBM cell growth in vitro. Xenograft tumor animal model was established to elucidate the in vivo function of miR-186-5p. MiR-186-5p directly targeted mRNAs of both FGF2 and RelA, and repressed their expressions. Invasive and migratory abilities of GBM cells and growth of xenograft tumors were significantly inhibited by miR-186-5p, which can be restored by re-introduction of FGF2 and RelA expressions. MiR-186-5p is a novel tumor suppressor miR that functions to inhibit tumorigenesis of GBM both in vitro and in vivo, by targeting both FGF2 and RelA. MiR-186-5p/FGF2/RelA pathway may be potentially used as molecular targets of in the clinical treatment of GBM.
Collapse
Affiliation(s)
- Fachen Wang
- Department of Neurosurgery, Yidu Central Hospital of Weifang, No. 4138 Linglongshan Road, Qingzhoushi, Shandong, People's Republic of China
| | - Hui Jiang
- Department of Medicine, Qingzhou Hospital of Traditional Chinese Medicine, No.2727 Haidai Road, Qingzhoushi, Shandong, People's Republic of China
| | - Shanjun Wang
- Department of Neurosurgery, Yidu Central Hospital of Weifang, No. 4138 Linglongshan Road, Qingzhoushi, Shandong, People's Republic of China
| | - Bing Chen
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, No. 1677 Wutaishan Road, Qingdao, Shandong, People's Republic of China.
| |
Collapse
|
23
|
Animal Models in Glioblastoma: Use in Biology and Developing Therapeutic Strategies. ADVANCES IN BIOLOGY AND TREATMENT OF GLIOBLASTOMA 2017. [DOI: 10.1007/978-3-319-56820-1_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
24
|
Henning RK, Varghese JO, Das S, Nag A, Tang G, Tang K, Sutherland AM, Heath JR. Degradation of Akt using protein-catalyzed capture agents. J Pept Sci 2016; 22:196-200. [PMID: 26880702 DOI: 10.1002/psc.2858] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/29/2015] [Accepted: 12/30/2015] [Indexed: 12/22/2022]
Abstract
Abnormal signaling of the protein kinase Akt has been shown to contribute to human diseases such as diabetes and cancer, but Akt has proven to be a challenging target for drugging. Using iterative in situ click chemistry, we recently developed multiple protein-catalyzed capture (PCC) agents that allosterically modulate Akt enzymatic activity in a protein-based assay. Here, we utilize similar PCCs to exploit endogenous protein degradation pathways. We use the modularity of the anti-Akt PCCs to prepare proteolysis targeting chimeric molecules that are shown to promote the rapid degradation of Akt in live cancer cells. These novel proteolysis targeting chimeric molecules demonstrate that the epitope targeting selectivity of PCCs can be coupled with non-traditional drugging moieties to inhibit challenging targets.
Collapse
Affiliation(s)
- Ryan K Henning
- Division of Chemistry and Chemical Engineering, California Institute of Technology, MC 127-72, Pasadena, CA 91125
| | - Joseph O Varghese
- Division of Chemistry and Chemical Engineering, California Institute of Technology, MC 127-72, Pasadena, CA 91125
| | - Samir Das
- Division of Chemistry and Chemical Engineering, California Institute of Technology, MC 127-72, Pasadena, CA 91125
| | - Arundhati Nag
- Division of Chemistry and Chemical Engineering, California Institute of Technology, MC 127-72, Pasadena, CA 91125
| | - Grace Tang
- Division of Chemistry and Chemical Engineering, California Institute of Technology, MC 127-72, Pasadena, CA 91125
| | - Kevin Tang
- Division of Chemistry and Chemical Engineering, California Institute of Technology, MC 127-72, Pasadena, CA 91125
| | - Alexander M Sutherland
- Division of Chemistry and Chemical Engineering, California Institute of Technology, MC 127-72, Pasadena, CA 91125
| | - James R Heath
- Division of Chemistry and Chemical Engineering, California Institute of Technology, MC 127-72, Pasadena, CA 91125
| |
Collapse
|
25
|
Thorek DLJ, Kramer RM, Chen Q, Jeong J, Lupu ME, Lee AM, Moynahan ME, Lowery M, Ulmert D, Zanzonico P, Deasy JO, Humm JL, Russell J. Reverse-Contrast Imaging and Targeted Radiation Therapy of Advanced Pancreatic Cancer Models. Int J Radiat Oncol Biol Phys 2015; 93:444-53. [PMID: 26238952 PMCID: PMC4575601 DOI: 10.1016/j.ijrobp.2015.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 05/26/2015] [Accepted: 06/01/2015] [Indexed: 01/23/2023]
Abstract
PURPOSE To evaluate the feasibility of delivering experimental radiation therapy to tumors in the mouse pancreas. Imaging and treatment were performed using combined CT (computed tomography)/orthovoltage treatment with a rotating gantry. METHODS AND MATERIALS After intraperitoneal administration of radiopaque iodinated contrast, abdominal organ delineation was performed by x-ray CT. With this technique we delineated the pancreas and both orthotopic xenografts and genetically engineered disease. Computed tomographic imaging was validated by comparison with magnetic resonance imaging. Therapeutic radiation was delivered via a 1-cm diameter field. Selective x-ray radiation therapy of the noninvasively defined orthotopic mass was confirmed using γH2AX staining. Mice could tolerate a dose of 15 Gy when the field was centered on the pancreas tail, and treatment was delivered as a continuous 360° arc. This strategy was then used for radiation therapy planning for selective delivery of therapeutic x-ray radiation therapy to orthotopic tumors. RESULTS Tumor growth delay after 15 Gy was monitored, using CT and ultrasound to determine the tumor volume at various times after treatment. Our strategy enables the use of clinical radiation oncology approaches to treat experimental tumors in the pancreas of small animals for the first time. We demonstrate that delivery of 15 Gy from a rotating gantry minimizes background healthy tissue damage and significantly retards tumor growth. CONCLUSIONS This advance permits evaluation of radiation planning and dosing parameters. Accurate noninvasive longitudinal imaging and monitoring of tumor progression and therapeutic response in preclinical models is now possible and can be expected to more effectively evaluate pancreatic cancer disease and therapeutic response.
Collapse
Affiliation(s)
- Daniel L J Thorek
- Division of Nuclear Medicine, The Russell H. Morgan Department of Radiology and Radiological Sciences, The Johns Hopkins School of Medicine, Baltimore, MD.
| | - Robin M Kramer
- Tri-Institutional Training Program in Laboratory Animal Medicine and Science, Memorial Sloan-Kettering Cancer Center (MSKCC), Weill Cornell Medical College, The Rockefeller University, New York, NY
| | - Qing Chen
- Department of Medical Physics, MSKCC, New York, NY
| | - Jeho Jeong
- Department of Medical Physics, MSKCC, New York, NY
| | | | | | | | | | - David Ulmert
- Molecular Pharmacology and Chemistry Program, MSKCC, New York, NY; Department of Surgery (Urology), Skåne University Hospital, Malmö, Sweden
| | | | | | - John L Humm
- Department of Medical Physics, MSKCC, New York, NY
| | | |
Collapse
|
26
|
Endaya BB, Lam PYP, Meedeniya ACB, Neuzil J. Transcriptional profiling of dividing tumor cells detects intratumor heterogeneity linked to cell proliferation in a brain tumor model. Mol Oncol 2015; 10:126-37. [PMID: 26388584 DOI: 10.1016/j.molonc.2015.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/11/2015] [Accepted: 09/01/2015] [Indexed: 01/02/2023] Open
Abstract
Intratumor heterogeneity is a primary feature of high-grade gliomas, complicating their therapy. As accumulating evidence suggests that intratumor heterogeneity is a consequence of cellular subsets with different cycling frequencies, we developed a method for transcriptional profiling of gliomas, using a novel technique to dissect the tumors into two fundamental cellular subsets, namely, the proliferating and non-proliferating cell fractions. The tumor fractions were sorted whilst maintaining their molecular integrity, by incorporating the thymidine analog 5-ethynyl-2'-deoxyuridine into actively dividing cells. We sorted the actively dividing versus non-dividing cells from cultured glioma cells, and parental and clonally derived orthotopic tumors, and analyzed them for a number of transcripts. While there was no significant difference in the transcriptional profiles between the two cellular subsets in cultured glioma cells, we demonstrate ∼2-6 fold increase in transcripts of cancer and neuronal stem cell and tumor cell migration/invasion markers, and ∼2-fold decrease in transcripts of markers of hypoxia and their target genes, in the dividing tumor cells of the orthotopic glioma when compared to their non-proliferative counterparts. This suggests the influence of the brain microenvironment in transcriptional regulation and, thereby, the physiology of glioma cells in vivo. When clonal glioma cells were derived from a parental glioma and the resultant orthotopic tumors were compared, their transcriptional profiles were closely correlated to tumor aggression and consequently, survival of the experimental animals. This study demonstrates the resolution of intratumor heterogeneity for profiling studies based on cell proliferation, a defining feature of cancers, with implications for treatment design.
Collapse
Affiliation(s)
- Berwini B Endaya
- School of Medical Science, Griffith University, Southport, Qld, 4222, Australia
| | - Paula Y P Lam
- Laboratory of Cancer Gene Therapy, National Cancer Centre, 169610, Singapore
| | - Adrian C B Meedeniya
- School of Medical Science, Griffith University, Southport, Qld, 4222, Australia.
| | - Jiri Neuzil
- School of Medical Science, Griffith University, Southport, Qld, 4222, Australia; Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague, 142 20, Czech Republic.
| |
Collapse
|
27
|
Pisco AO, Huang S. Non-genetic cancer cell plasticity and therapy-induced stemness in tumour relapse: 'What does not kill me strengthens me'. Br J Cancer 2015; 112:1725-32. [PMID: 25965164 PMCID: PMC4647245 DOI: 10.1038/bjc.2015.146] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 03/17/2015] [Accepted: 03/23/2015] [Indexed: 12/16/2022] Open
Abstract
Therapy resistance and tumour relapse after drug therapy are commonly explained by Darwinian selection of pre-existing drug-resistant, often stem-like cancer cells resulting from random mutations. However, the ubiquitous non-genetic heterogeneity and plasticity of tumour cell phenotype raises the question: are mutations really necessary and sufficient to promote cell phenotype changes during tumour progression? Cancer therapy inevitably spares some cancer cells, even in the absence of resistant mutants. Accumulating observations suggest that the non-killed, residual tumour cells actively acquire a new phenotype simply by exploiting their developmental potential. These surviving cells are stressed by the cytotoxic treatment, and owing to phenotype plasticity, exhibit a variety of responses. Some are pushed into nearby, latent attractor states of the gene regulatory network which resemble evolutionary ancient or early developmental gene expression programs that confer stemness and resilience. By entering such stem-like, stress-response states, the surviving cells strengthen their capacity to cope with future noxious agents. Considering non-genetic cell state dynamics and the relative ease with which surviving but stressed cells can be tipped into latent attractors provides a foundation for exploring new therapeutic approaches that seek not only to kill cancer cells but also to avoid promoting resistance and relapse that are inherently linked to the attempts to kill them.
Collapse
Affiliation(s)
- A O Pisco
- 1] Institute for Systems Biology, Seattle, WA 98109, USA [2] Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - S Huang
- 1] Institute for Systems Biology, Seattle, WA 98109, USA [2] Institute for Biocomplexity and Informatics, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
28
|
Abstract
A number of agents are used clinically to enhance the efficacy of radiotherapy today, many of which are cytotoxic chemotherapies. Agents that enhance radiation induced tumor cell killing or protect normal tissues from the deleterious effects of ionizing radiation are collectively termed radiation modifiers. A significant effort in radiobiological research is geared towards describing and testing radiation modifiers with the intent of enhancing the therapeutic effects of radiation while minimizing normal tissue toxicity. In this review, we discuss the characteristics of these agents, the testing required to translate these agents into clinical trials, and highlight some challenges in these efforts.
Collapse
Affiliation(s)
- Deborah E Citrin
- Radiation Oncology Branch and Radiation Biology Branch of the National Cancer Institute, Bethesda, MD.
| | - James B Mitchell
- Radiation Oncology Branch and Radiation Biology Branch of the National Cancer Institute, Bethesda, MD
| |
Collapse
|
29
|
McNeill RS, Vitucci M, Wu J, Miller CR. Contemporary murine models in preclinical astrocytoma drug development. Neuro Oncol 2014; 17:12-28. [PMID: 25246428 DOI: 10.1093/neuonc/nou288] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Despite 6 decades of research, only 3 drugs have been approved for astrocytomas, the most common malignant primary brain tumors. However, clinical drug development is accelerating with the transition from empirical, cytotoxic therapy to precision, targeted medicine. Preclinical animal model studies are critical for prioritizing drug candidates for clinical development and, ultimately, for their regulatory approval. For decades, only murine models with established tumor cell lines were available for such studies. However, these poorly represent the genomic and biological properties of human astrocytomas, and their preclinical use fails to accurately predict efficacy in clinical trials. Newer models developed over the last 2 decades, including patient-derived xenografts, genetically engineered mice, and genetically engineered cells purified from human brains, more faithfully phenocopy the genomics and biology of human astrocytomas. Harnessing the unique benefits of these models will be required to identify drug targets, define combination therapies that circumvent inherent and acquired resistance mechanisms, and develop molecular biomarkers predictive of drug response and resistance. With increasing recognition of the molecular heterogeneity of astrocytomas, employing multiple, contemporary models in preclinical drug studies promises to increase the efficiency of drug development for specific, molecularly defined subsets of tumors.
Collapse
Affiliation(s)
- Robert S McNeill
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina (R.S.M., M.V., C.R.M.); Departments of Neurosurgery and Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina (J.W.); Department of Neurology, Lineberger Comprehensive Cancer Center, and Neurosciences Center University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M.)
| | - Mark Vitucci
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina (R.S.M., M.V., C.R.M.); Departments of Neurosurgery and Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina (J.W.); Department of Neurology, Lineberger Comprehensive Cancer Center, and Neurosciences Center University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M.)
| | - Jing Wu
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina (R.S.M., M.V., C.R.M.); Departments of Neurosurgery and Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina (J.W.); Department of Neurology, Lineberger Comprehensive Cancer Center, and Neurosciences Center University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M.)
| | - C Ryan Miller
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina (R.S.M., M.V., C.R.M.); Departments of Neurosurgery and Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina (J.W.); Department of Neurology, Lineberger Comprehensive Cancer Center, and Neurosciences Center University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M.)
| |
Collapse
|
30
|
Cohen JT, Gil Z, Binenbaum Y, Na'ara S, Amit M. An orthotopic mouse model of laryngeal squamous cell carcinoma. Ann Otol Rhinol Laryngol 2014; 124:143-7. [PMID: 25204712 DOI: 10.1177/0003489414549575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE This study aimed to create a reliable and reproducible orthotopic mouse model of laryngeal malignancy that recapitulates its biologic behavior, local invasiveness, and spread as seen in patients. METHODS Via direct laryngoscopy, human squamous cell carcinoma line FaDu (ATCC HTB-43) was implanted in the larynx (supraglottis and glottis) in nu/nu mice (n = 31). Animals were monitored with direct laryngoscopy and ultrasound for tumor growth and survival. Specimens of larynxes, neck lymphatics, livers, and lungs were removed for histopathologic evaluation to assess tumor extension, thyroid cartilage invasion, and nodal spread. RESULTS Thirty-one successful direct laryngoscopies were performed. Supraglottic and glottic tumor uptake was 100% and 25%, respectively. Median survival for the animals with supraglottic tumors was 35 days. Histopathologic evaluation revealed pre-epiglottic extension, paraglottic extension, thyroid cartilage invasion, and lymph node metastasis. CONCLUSION We describe the first orthotopic model for laryngeal cancer. Our model faithfully recapitulates the phenotype and malignant behavior that reproduces its natural biologic behavior as seen in laryngeal cancer patients. This model offers an opportunity to identify and specifically target therapy for larynx squamous cell carcinoma.
Collapse
Affiliation(s)
- Jacob T Cohen
- Department of Otolaryngology-Head and Neck Surgery, Rambam Health Care Campus, Haifa, Israel
| | - Ziv Gil
- Department of Otolaryngology-Head and Neck Surgery, Rambam Health Care Campus, Haifa, Israel The Laboratory for Applied Cancer Research, Clinical Research Center at Rambam, Rambam Health Care Campus, The Technion, Israel Institute of Technology, Haifa, Israel
| | - Yoav Binenbaum
- The Laboratory for Applied Cancer Research, Clinical Research Center at Rambam, Rambam Health Care Campus, The Technion, Israel Institute of Technology, Haifa, Israel
| | - Shorook Na'ara
- Department of Otolaryngology-Head and Neck Surgery, Rambam Health Care Campus, Haifa, Israel The Laboratory for Applied Cancer Research, Clinical Research Center at Rambam, Rambam Health Care Campus, The Technion, Israel Institute of Technology, Haifa, Israel
| | - Moran Amit
- Department of Otolaryngology-Head and Neck Surgery, Rambam Health Care Campus, Haifa, Israel The Laboratory for Applied Cancer Research, Clinical Research Center at Rambam, Rambam Health Care Campus, The Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
31
|
Peng XC, Wang M, Chen XX, Liu J, Xiao GH, Liao HL. Plasmid-encoding vasostatin inhibited the growth and metastasis of human hepatocellular carcinoma cells. Mol Cell Biochem 2014; 395:265-72. [PMID: 24997628 DOI: 10.1007/s11010-014-2135-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 06/17/2014] [Indexed: 02/05/2023]
Abstract
The growth and metastasis of solid tumors depends on angiogenesis. Anti-angiogenesis therapy may represent a promising therapeutic option. Vasostatin, the N-terminal domain of calreticulin, is a very potent endogenous inhibitor of angiogenesis and tumor growth. In this study, we attempted to investigate whether plasmid-encoding vasostatin complexed with cationic liposome could suppress the growth and metastasis of hepatocellular carcinoma in vivo and discover its possible mechanism of action. Apoptosis induction of pSecTag2B-vasostatin plasmid on murine endothelial cells (MS1) was examined by flow cytometric analysis in vitro. Nude mice bearing HCCLM3 tumor received pSecTag2B-vasostatin, pSecTag2B-Null, and 0.9 % NaCl solution, respectively. Tumor net weight was measured and survival time was observed. Microvessel density within tumor tissues was determined by CD31 immunohistochemistry. H&E staining of lungs and TUNEL assay of primary tumor tissues were also conducted. The results displayed that pSecTag2B-vasostatin could inhibit the growth and metastasis of hepatocellular carcinoma xenografts and prolong survival time compared with the controls in vivo. Moreover, histologic analysis revealed that pSecTag2B-vasostatin treatment increased apoptosis and inhibited angiogenesis. The present data may be of importance to the further exploration of this new anti-angiogenesis approach in the treatment of hepatocellular cancer.
Collapse
Affiliation(s)
- Xing-Chen Peng
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China,
| | | | | | | | | | | |
Collapse
|
32
|
Johnson H, White FM. Quantitative analysis of signaling networks across differentially embedded tumors highlights interpatient heterogeneity in human glioblastoma. J Proteome Res 2014; 13:4581-93. [PMID: 24927040 PMCID: PMC4227552 DOI: 10.1021/pr500418w] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
Glioblastoma
multiforme (GBM) is the most aggressive malignant
primary brain tumor, with a dismal mean survival even with the current
standard of care. Although in vitro cell systems can provide mechanistic
insight into the regulatory networks governing GBM cell proliferation
and migration, clinical samples provide a more physiologically relevant
view of oncogenic signaling networks. However, clinical samples are
not widely available and may be embedded for histopathologic analysis.
With the goal of accurately identifying activated signaling networks
in GBM tumor samples, we investigated the impact of embedding in optimal
cutting temperature (OCT) compound followed by flash freezing in LN2 vs immediate flash freezing (iFF) in LN2 on protein
expression and phosphorylation-mediated signaling networks. Quantitative
proteomic and phosphoproteomic analysis of 8 pairs of tumor specimens
revealed minimal impact of the different sample processing strategies
and highlighted the large interpatient heterogeneity present in these
tumors. Correlation analyses of the differentially processed tumor
sections identified activated signaling networks present in selected
tumors and revealed the differential expression of transcription,
translation, and degradation associated proteins. This study demonstrates
the capability of quantitative mass spectrometry for identification
of in vivo oncogenic signaling networks from human tumor specimens
that were either OCT-embedded or immediately flash-frozen.
Collapse
Affiliation(s)
- Hannah Johnson
- Department of Biological Engineering, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | | |
Collapse
|
33
|
Lee BJ, Kim JH, Jo DH, Kim KW, Yu YS, Kim JH. Nuclear expression of p53 in mature tumor endothelium of retinoblastoma. Oncol Rep 2014; 32:801-7. [PMID: 24898002 DOI: 10.3892/or.2014.3236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 05/16/2014] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the p53 expression pattern in tumor cells and in mature tumor vascular endothelium of retinoblastoma. Nuclear p53 accumulation was observed in most of the tumor cells in both the human and orthotopic retinoblastoma animal models using SNUOT-Rb1 and Y79 cells. In the orthotopic animal model, some of the tumor vascular endothelium also demonstrated nuclear p53 immunoreactivity, and the ratio of p53 positivity among the total mature tumor vascular endothelium was slightly higher in the Y79 cell model when compared with the SNUOT-Rb1 cell model. In addition, in the human retinoblastoma specimens, 32.9% of the tumor vascular endothelium showed p53 nuclear staining. In conclusion, some of the mature tumor vascular endothelium in both the human and orthotopic models of retinoblastoma share the same cytogenetic abnormality (an abnormal nuclear accumulation of p53) with retinoblastoma cells.
Collapse
Affiliation(s)
- Byung Joo Lee
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jin Hyoung Kim
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong Hyun Jo
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyu-Won Kim
- Tumor Microenvironment Research Center, Global Core Research Center, Seoul National University, Seoul, Republic of Korea
| | - Young Suk Yu
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jeong Hun Kim
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
34
|
Hollingshead MG, Stockwin LH, Alcoser SY, Newton DL, Orsburn BC, Bonomi CA, Borgel SD, Divelbiss R, Dougherty KM, Hager EJ, Holbeck SL, Kaur G, Kimmel DJ, Kunkel MW, Millione A, Mullendore ME, Stotler H, Collins J. Gene expression profiling of 49 human tumor xenografts from in vitro culture through multiple in vivo passages--strategies for data mining in support of therapeutic studies. BMC Genomics 2014; 15:393. [PMID: 24885658 PMCID: PMC4041995 DOI: 10.1186/1471-2164-15-393] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 05/09/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Development of cancer therapeutics partially depends upon selection of appropriate animal models. Therefore, improvements to model selection are beneficial. RESULTS Forty-nine human tumor xenografts at in vivo passages 1, 4 and 10 were subjected to cDNA microarray analysis yielding a dataset of 823 Affymetrix HG-U133 Plus 2.0 arrays. To illustrate mining strategies supporting therapeutic studies, transcript expression was determined: 1) relative to other models, 2) with successive in vivo passage, and 3) during the in vitro to in vivo transition. Ranking models according to relative transcript expression in vivo has the potential to improve initial model selection. For example, combining p53 tumor expression data with mutational status could guide selection of tumors for therapeutic studies of agents where p53 status purportedly affects efficacy (e.g., MK-1775). The utility of monitoring changes in gene expression with extended in vivo tumor passages was illustrated by focused studies of drug resistance mediators and receptor tyrosine kinases. Noteworthy observations included a significant decline in HCT-15 colon xenograft ABCB1 transporter expression and increased expression of the kinase KIT in A549 with serial passage. These trends predict sensitivity to agents such as paclitaxel (ABCB1 substrate) and imatinib (c-KIT inhibitor) would be altered with extended passage. Given that gene expression results indicated some models undergo profound changes with in vivo passage, a general metric of stability was generated so models could be ranked accordingly. Lastly, changes occurring during transition from in vitro to in vivo growth may have important consequences for therapeutic studies since targets identified in vitro could be over- or under-represented when tumor cells adapt to in vivo growth. A comprehensive list of mouse transcripts capable of cross-hybridizing with human probe sets on the HG-U133 Plus 2.0 array was generated. Removal of the murine artifacts followed by pairwise analysis of in vitro cells with respective passage 1 xenografts and GO analysis illustrates the complex interplay that each model has with the host microenvironment. CONCLUSIONS This study provides strategies to aid selection of xenograft models for therapeutic studies. These data highlight the dynamic nature of xenograft models and emphasize the importance of maintaining passage consistency throughout experiments.
Collapse
Affiliation(s)
- Melinda G Hollingshead
- />Biological Testing Branch, National Cancer Institute at Frederick, 1050 Boyles Street, Building 1043, Room 11, Frederick, MD 21702 USA
| | - Luke H Stockwin
- />Biological Testing Branch, Developmental Therapeutics Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Sergio Y Alcoser
- />Biological Testing Branch, Developmental Therapeutics Program, National Cancer Institute at Frederick, Frederick, MD 21702 USA
| | - Dianne L Newton
- />Biological Testing Branch, Developmental Therapeutics Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | | | - Carrie A Bonomi
- />Biological Testing Branch, Developmental Therapeutics Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Suzanne D Borgel
- />Biological Testing Branch, Developmental Therapeutics Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Raymond Divelbiss
- />Biological Testing Branch, Developmental Therapeutics Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Kelly M Dougherty
- />Biological Testing Branch, Developmental Therapeutics Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Elizabeth J Hager
- />Biological Testing Branch, Developmental Therapeutics Program, National Cancer Institute at Frederick, Frederick, MD 21702 USA
| | - Susan L Holbeck
- />Information Technology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, MD 20892 USA
| | - Gurmeet Kaur
- />Molecular Pharmacology Branch, Developmental Therapeutics Program, National Cancer Institute at Frederick, Frederick, MD 21702 USA
| | - David J Kimmel
- />Biological Testing Branch, Developmental Therapeutics Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Mark W Kunkel
- />Information Technology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, MD 20892 USA
| | - Angelena Millione
- />Biological Testing Branch, Developmental Therapeutics Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Michael E Mullendore
- />Biological Testing Branch, Developmental Therapeutics Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Howard Stotler
- />Biological Testing Branch, Developmental Therapeutics Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Jerry Collins
- />Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, MD 20892 USA
| |
Collapse
|
35
|
Pilgaard L, Mortensen JH, Henriksen M, Olesen P, Sørensen P, Laursen R, Vyberg M, Agger R, Zachar V, Moos T, Duroux M. Cripto-1 expression in glioblastoma multiforme. Brain Pathol 2014; 24:360-70. [PMID: 24521322 DOI: 10.1111/bpa.12131] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 02/05/2014] [Indexed: 01/24/2023] Open
Abstract
Human glioblastoma multiforme (GBM) is an aggressive cancer with a very poor prognosis. Cripto-1 (CR-1) has a key regulatory role in embryogenesis, while in adult tissue re-expression of CR-1 has been correlated to malignant progression in solid cancers of non-neuronal origin. As CR-1 expression has yet to be described in cerebral cancer and CR-1 is regulated by signaling pathways dysregulated in GBM, we aimed to investigate CR-1 in the context of expression in GBM. The study was performed using enzyme-linked immunosorbent assay (ELISA), Western blotting, polymerase chain reaction (PCR) and immunohistochemistry to analyze the blood and tissue from 28 GBM and 4 low-grade glioma patients. Within the patient cohort, we found high CR-1 protein levels in blood plasma to significantly correlate with a shorter overall survival. We identified CR-1 in different areas of GBM tissue, including perivascular tumor cells, and in endothelial cells. Collectively, our data suggest that CR-1 could be a prognostic biomarker for GBM with the potential of being a therapeutic target.
Collapse
Affiliation(s)
- Linda Pilgaard
- Laboratory of Cancer Biology, Biomedicine Group, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ionizing radiation and glioblastoma exosomes: implications in tumor biology and cell migration. Transl Oncol 2013; 6:638-48. [PMID: 24466366 DOI: 10.1593/tlo.13640] [Citation(s) in RCA: 183] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 09/25/2013] [Accepted: 10/30/2013] [Indexed: 12/12/2022] Open
Abstract
Exosomes are nanometer-sized lipid vesicles released ubiquitously by cells, which have been shown to have a normal physiological role, as well as influence the tumor microenvironment and aid metastasis. Recent studies highlight the ability of exosomes to convey tumor-suppressive and oncogenic mRNAs, microRNAs, and proteins to a receiving cell, subsequently activating downstream signaling pathways and influencing cellular phenotype. Here, we show that radiation increases the abundance of exosomes released by glioblastoma cells and normal astrocytes. Exosomes derived from irradiated cells enhanced the migration of recipient cells, and their molecular profiling revealed an abundance of molecules related to signaling pathways important for cell migration. In particular, connective tissue growth factor (CTGF) mRNA and insulin-like growth factor binding protein 2 (IGFBP2) protein levels were elevated, and coculture of nonirradiated cells with exosomes isolated from irradiated cells increased CTGF protein expression in the recipient cells. Additionally, these exosomes enhanced the activation of neurotrophic tyrosine kinase receptor type 1 (TrkA), focal adhesion kinase, Paxillin, and proto-oncogene tyrosine-protein kinase Src (Src) in recipient cells, molecules involved in cell migration. Collectively, our data suggest that radiation influences exosome abundance, specifically alters their molecular composition, and on uptake, promotes a migratory phenotype.
Collapse
|
37
|
Jin H, Liang L, Liu L, Deng W, Liu J. HDAC inhibitor DWP0016 activates p53 transcription and acetylation to inhibit cell growth in U251 glioblastoma cells. J Cell Biochem 2013; 114:1498-509. [PMID: 23297003 DOI: 10.1002/jcb.24491] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 12/18/2012] [Indexed: 11/11/2022]
Abstract
Here we report a hydroacid named DWP0016, which exhibited HDAC inhibition and induced p53 acetylation in U251 glioblastoma cells. DWP0016 effectively inhibited the cell growth of U251 cells and other four carcinoma cell lines but did not affect the normal cells. Cell cycle distribution analysis showed DWP0016 arrested at G1 phase cell cycle dose-dependently in U251 cells. DWP0016 induced caspase-dependent and independent apoptosis in U251 cells, which was identified by flow cytometry analysis, caspases activity analysis, Western blotting assay, and caspases inhibition. Mechanisms research suggested that DWP0016 activated transcription and acetylation of tumor suppressor p53. DWP0016 regulated p300, CBP, and PCAF to facilitate p53 acetylation at lys382 in U251 cells. In addition, activation of p53 by DWP0016 promoted PUMA to catalyze mitochondrial pathway. Besides, siRNA assay indicated p53 was the key gene to induce growth inhibition, cell cycle arrest, and apoptosis in DWP0016 treated U251 cells. Conclusively, our results show DWP0016 is a potent HDAC inhibitor and the anti-tumor activity is consistent with its intended p53 activation mechanisms. These findings indicate the promising antitumor potential of DWP0016 for further glioblastoma treatment applications.
Collapse
Affiliation(s)
- Hui Jin
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, People's Republic of China
| | | | | | | | | |
Collapse
|
38
|
Yost SE, Pastorino S, Rozenzhak S, Smith EN, Chao YS, Jiang P, Kesari S, Frazer KA, Harismendy O. High-resolution mutational profiling suggests the genetic validity of glioblastoma patient-derived pre-clinical models. PLoS One 2013; 8:e56185. [PMID: 23441165 PMCID: PMC3575368 DOI: 10.1371/journal.pone.0056185] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 01/07/2013] [Indexed: 11/19/2022] Open
Abstract
Recent advances in the ability to efficiently characterize tumor genomes is enabling targeted drug development, which requires rigorous biomarker-based patient selection to increase effectiveness. Consequently, representative DNA biomarkers become equally important in pre-clinical studies. However, it is still unclear how well these markers are maintained between the primary tumor and the patient-derived tumor models. Here, we report the comprehensive identification of somatic coding mutations and copy number aberrations in four glioblastoma (GBM) primary tumors and their matched pre-clinical models: serum-free neurospheres, adherent cell cultures, and mouse xenografts. We developed innovative methods to improve the data quality and allow a strict comparison of matched tumor samples. Our analysis identifies known GBM mutations altering PTEN and TP53 genes, and new actionable mutations such as the loss of PIK3R1, and reveals clear patient-to-patient differences. In contrast, for each patient, we do not observe any significant remodeling of the mutational profile between primary to model tumors and the few discrepancies can be attributed to stochastic errors or differences in sample purity. Similarly, we observe ∼96% primary-to-model concordance in copy number calls in the high-cellularity samples. In contrast to previous reports based on gene expression profiles, we do not observe significant differences at the DNA level between in vitro compared to in vivo models. This study suggests, at a remarkable resolution, the genome-wide conservation of a patient’s tumor genetics in various pre-clinical models, and therefore supports their use for the development and testing of personalized targeted therapies.
Collapse
Affiliation(s)
- Shawn E. Yost
- Bioinformatics and Systems Biology Graduate Program, University of California San Diego, La Jolla, California, United States of America
- Division of Genome Information Sciences, Department of Pediatrics and Rady Children’s Hospital, University of California San Diego, La Jolla, California, United States of America
| | - Sandra Pastorino
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Translational Neuro-oncology Laboratories, University of California San Diego, La Jolla, California, United States of America
| | - Sophie Rozenzhak
- Division of Genome Information Sciences, Department of Pediatrics and Rady Children’s Hospital, University of California San Diego, La Jolla, California, United States of America
| | - Erin N. Smith
- Division of Genome Information Sciences, Department of Pediatrics and Rady Children’s Hospital, University of California San Diego, La Jolla, California, United States of America
| | - Ying S. Chao
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Translational Neuro-oncology Laboratories, University of California San Diego, La Jolla, California, United States of America
| | - Pengfei Jiang
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Translational Neuro-oncology Laboratories, University of California San Diego, La Jolla, California, United States of America
| | - Santosh Kesari
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Translational Neuro-oncology Laboratories, University of California San Diego, La Jolla, California, United States of America
- Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, United States of America
- * E-mail: (OH); (SK)
| | - Kelly A. Frazer
- Division of Genome Information Sciences, Department of Pediatrics and Rady Children’s Hospital, University of California San Diego, La Jolla, California, United States of America
- Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, United States of America
- Clinical and Translational Research Institute, University of California San Diego, La Jolla, California, United States of America
- Institute for Genomic Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Olivier Harismendy
- Division of Genome Information Sciences, Department of Pediatrics and Rady Children’s Hospital, University of California San Diego, La Jolla, California, United States of America
- Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, United States of America
- Clinical and Translational Research Institute, University of California San Diego, La Jolla, California, United States of America
- * E-mail: (OH); (SK)
| |
Collapse
|
39
|
Chernet B, Levin M. Endogenous Voltage Potentials and the Microenvironment: Bioelectric Signals that Reveal, Induce and Normalize Cancer. JOURNAL OF CLINICAL & EXPERIMENTAL ONCOLOGY 2013; Suppl 1:S1-002. [PMID: 25525610 PMCID: PMC4267524 DOI: 10.4172/2324-9110.s1-002] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cancer may be a disease of geometry: a misregulation of the field of information that orchestrates individual cells' activities towards normal anatomy. Recent work identified molecular mechanisms underlying a novel system of developmental control: bioelectric gradients. Endogenous spatio-temporal differences in resting potential of non-neural cells provide instructive cues for cell regulation and complex patterning during embryogenesis and regeneration. It is now appreciated that these cues are an important layer of the dysregulation of cell: cell interactions that leads to cancer. Abnormal depolarization of resting potential (Vmem) is a convenient marker for neoplasia and activates a metastatic phenotype in genetically-normal cells in vivo. Moreover, oncogene expression depolarizes cells that form tumor-like structures, but is unable to form tumors if this depolarization is artificially prevented by misexpression of hyperpolarizing ion channels. Vmem triggers metastatic behaviors at considerable distance, mediated by transcriptional and epigenetic effects of electrically-modulated flows of serotonin and butyrate. While in vivo data on voltages in carcinogenesis comes mainly from the amphibian model, unbiased genetic screens and network profiling in rodents and human tissues reveal several ion channel proteins as bona fide oncogene and promising targets for cancer drug development. However, we propose that a focus on specific channel genes is just the tip of the iceberg. Bioelectric state is determined by post-translational gating of ion channels, not only from genetically-specified complements of ion translocators. A better model is a statistical dynamics view of spatial Vmem gradients. Cancer may not originate at the single cell level, since gap junctional coupling results in multi-cellular physiological networks with multiple stable attractors in bioelectrical state space. New medical applications await a detailed understanding of the mechanisms by which organ target morphology stored in real-time patterns of ion flows is perceived or mis-perceived by cells. Mastery of somatic voltage gradients will lead to cancer normalization or rebooting strategies, such as those that occur in regenerating and embryonic organs, resulting in transformative advances in basic biology and oncology.
Collapse
Affiliation(s)
| | - Michael Levin
- Corresponding author: Michael Levin, Department of Biology, Tufts Center for Regenerative and Developmental Biology, Tufts University, 200 Boston Ave., Suite 4600, Medford, MA 02155, USA, Tel: (617) 627-6161; Fax:(617) 627- 6121;
| |
Collapse
|
40
|
Kahn J, Tofilon PJ, Camphausen K. Preclinical models in radiation oncology. Radiat Oncol 2012; 7:223. [PMID: 23270380 PMCID: PMC3549821 DOI: 10.1186/1748-717x-7-223] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 12/18/2012] [Indexed: 11/10/2022] Open
Abstract
As the incidence of cancer continues to rise, the use of radiotherapy has emerged as a leading treatment modality. Preclinical models in radiation oncology are essential tools for cancer research and therapeutics. Various model systems have been used to test radiation therapy, including in vitro cell culture assays as well as in vivo ectopic and orthotopic xenograft models. This review aims to describe such models, their advantages and disadvantages, particularly as they have been employed in the discovery of molecular targets for tumor radiosensitization. Ultimately, any model system must be judged by its utility in developing more effective cancer therapies, which is in turn dependent on its ability to simulate the biology of tumors as they exist in situ. Although every model has its limitations, each has played a significant role in preclinical testing. Continued advances in preclinical models will allow for the identification and application of targets for radiation in the clinic.
Collapse
Affiliation(s)
- Jenna Kahn
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
41
|
Resistance to two heterologous neurotropic oncolytic viruses, Semliki Forest virus and vaccinia virus, in experimental glioma. J Virol 2012; 87:2363-6. [PMID: 23221568 DOI: 10.1128/jvi.01609-12] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Attenuated Semliki Forest virus (SFV) may be suitable for targeting malignant glioma due to its natural neurotropism, but its replication in brain tumor cells may be restricted by innate antiviral defenses. We attempted to facilitate SFV replication in glioma cells by combining it with vaccinia virus, which is capable of antagonizing such defenses. Surprisingly, we found parenchymal mouse brain tumors to be refractory to both viruses. Also, vaccinia virus appears to be sensitive to SFV-induced antiviral interference.
Collapse
|
42
|
Shankavaram UT, Bredel M, Burgan WE, Carter D, Tofilon P, Camphausen K. Molecular profiling indicates orthotopic xenograft of glioma cell lines simulate a subclass of human glioblastoma. J Cell Mol Med 2012; 16:545-54. [PMID: 21595825 PMCID: PMC3164941 DOI: 10.1111/j.1582-4934.2011.01345.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Cell line models have been widely used to investigate glioblastoma multiforme (GBM) pathobiology and in the development of targeted therapies. However, GBM tumours are molecularly heterogeneous and how cell lines can best model that diversity is unknown. In this report, we investigated gene expression profiles of three preclinical growth models of glioma cell lines, in vitro and in vivo as subcutaneous and intracerebral xenografts to examine which cell line model most resembles the clinical samples. Whole genome DNA microarrays were used to profile gene expression in a collection of 25 high-grade glioblastomas, and comparisons were made to profiles of cell lines under three different growth models. Hierarchical clustering revealed three molecular subtypes of the glioblastoma patient samples. Supervised learning algorithm, trained on glioma subtypes predicted the intracerebral cell line model with one glioma subtype (r = 0.68; 95% bootstrap CI –0.41, 0.46). Survival analysis of enriched gene sets (P < 0.05) revealed 19 biological categories (146 genes) belonging to neuronal, signal transduction, apoptosis- and glutamate-mediated neurotransmitter activation signals that are associated with poor prognosis in this glioma subclass. We validated the expression profiles of these gene categories in an independent cohort of patients from ‘The Cancer Genome Atlas’ project (r = 0.62, 95% bootstrap CI: –0.42, 0.43). We then used these data to select and inhibit a novel target (glutamate receptor) and showed that LY341595, a glutamate receptor specific antagonist, could prolong survival in intracerebral tumour-implanted mice in combination with irradiation, providing an in vivo cell line system of preclinical studies.
Collapse
Affiliation(s)
- Uma T Shankavaram
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
43
|
Blakeley J, Grossman SA. Chemotherapy with cytotoxic and cytostatic agents in brain cancer. HANDBOOK OF CLINICAL NEUROLOGY 2012; 104:229-54. [PMID: 22230447 DOI: 10.1016/b978-0-444-52138-5.00017-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
44
|
Abstract
GBM (glioblastoma multiforme) is a highly aggressive brain tumour with very poor prognosis despite multi-modalities of treatment. Furthermore, recent failure of targeted therapy for these tumours highlights the need of appropriate rodent models for preclinical studies. In this review, we highlight the most commonly used rodent models (U251, U86, GL261, C6, 9L and CNS-1) with a focus on the pathological and genetic similarities to the human disease. We end with a comprehensive review of the CNS-1 rodent model.
Collapse
|
45
|
Wirth H, Löffler M, von Bergen M, Binder H. Expression cartography of human tissues using self organizing maps. BMC Bioinformatics 2011; 12:306. [PMID: 21794127 PMCID: PMC3161046 DOI: 10.1186/1471-2105-12-306] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 07/27/2011] [Indexed: 12/02/2022] Open
Abstract
Background Parallel high-throughput microarray and sequencing experiments produce vast quantities of multidimensional data which must be arranged and analyzed in a concerted way. One approach to addressing this challenge is the machine learning technique known as self organizing maps (SOMs). SOMs enable a parallel sample- and gene-centered view of genomic data combined with strong visualization and second-level analysis capabilities. The paper aims at bridging the gap between the potency of SOM-machine learning to reduce dimension of high-dimensional data on one hand and practical applications with special emphasis on gene expression analysis on the other hand. Results The method was applied to generate a SOM characterizing the whole genome expression profiles of 67 healthy human tissues selected from ten tissue categories (adipose, endocrine, homeostasis, digestion, exocrine, epithelium, sexual reproduction, muscle, immune system and nervous tissues). SOM mapping reduces the dimension of expression data from ten of thousands of genes to a few thousand metagenes, each representing a minicluster of co-regulated single genes. Tissue-specific and common properties shared between groups of tissues emerge as a handful of localized spots in the tissue maps collecting groups of co-regulated and co-expressed metagenes. The functional context of the spots was discovered using overrepresentation analysis with respect to pre-defined gene sets of known functional impact. We found that tissue related spots typically contain enriched populations of genes related to specific molecular processes in the respective tissue. Analysis techniques normally used at the gene-level such as two-way hierarchical clustering are better represented and provide better signal-to-noise ratios if applied to the metagenes. Metagene-based clustering analyses aggregate the tissues broadly into three clusters containing nervous, immune system and the remaining tissues. Conclusions The SOM technique provides a more intuitive and informative global view of the behavior of a few well-defined modules of correlated and differentially expressed genes than the separate discovery of the expression levels of hundreds or thousands of individual genes. The program is available as R-package 'oposSOM'.
Collapse
Affiliation(s)
- Henry Wirth
- Interdisciplinary Centre for Bioinformatics of Leipzig University, D-4107 Leipzig, Härtelstr. 16-18, Germany.
| | | | | | | |
Collapse
|
46
|
Wang X, Duan X, Yang G, Zhang X, Deng L, Zheng H, Deng C, Wen J, Wang N, Peng C, Zhao X, Wei Y, Chen L. Honokiol crosses BBB and BCSFB, and inhibits brain tumor growth in rat 9L intracerebral gliosarcoma model and human U251 xenograft glioma model. PLoS One 2011; 6:e18490. [PMID: 21559510 PMCID: PMC3084695 DOI: 10.1371/journal.pone.0018490] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 03/09/2011] [Indexed: 02/05/2023] Open
Abstract
Background Gliosarcoma is one of the most common malignant brain tumors, and anti-angiogenesis is a promising approach for the treatment of gliosarcoma. However, chemotherapy is obstructed by the physical obstacle formed by the blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB). Honokiol has been known to possess potent activities in the central nervous system diseases, and anti-angiogenic and anti-tumor properties. Here, we hypothesized that honokiol could cross the BBB and BCSFB for the treatment of gliosarcoma. Methodologies We first evaluated the abilities of honokiol to cross the BBB and BCSFB by measuring the penetration of honokiol into brain and blood-cerebrospinal fluid, and compared the honokiol amount taken up by brain with that by other tissues. Then we investigated the effect of honokiol on the growth inhibition of rat 9L gliosarcoma cells and human U251 glioma cells in vitro. Finally we established rat 9L intracerebral gliosarcoma model in Fisher 344 rats and human U251 xenograft glioma model in nude mice to investigate the anti-tumor activity. Principal Findings We showed for the first time that honokiol could effectively cross BBB and BCSFB. The ratios of brain/plasma concentration were respectively 1.29, 2.54, 2.56 and 2.72 at 5, 30, 60 and 120 min. And about 10% of honokiol in plasma crossed BCSFB into cerebrospinal fluid (CSF). In vitro, honokiol produced dose-dependent inhibition of the growth of rat 9L gliosarcoma cells and human U251 glioma cells with IC50 of 15.61 µg/mL and 16.38 µg/mL, respectively. In vivo, treatment with 20 mg/kg body weight of honokiol (honokiol was given twice per week for 3 weeks by intravenous injection) resulted in significant reduction of tumor volume (112.70±10.16 mm3) compared with vehicle group (238.63±19.69 mm3, P = 0.000), with 52.77% inhibiting rate in rat 9L intracerebral gliosarcoma model, and (1450.83±348.36 mm3) compared with vehicle group (2914.17±780.52 mm3, P = 0.002), with 50.21% inhibiting rate in human U251 xenograft glioma model. Honokiol also significantly improved the survival over vehicle group in the two models (P<0.05). Conclusions/Significance This study provided the first evidence that honokiol could effectively cross BBB and BCSFB and inhibit brain tumor growth in rat 9L intracerebral gliosarcoma model and human U251 xenograft glioma model. It suggested a significant strategy for offering a potential new therapy for the treatment of gliosarcoma.
Collapse
Affiliation(s)
- Xianhuo Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xingmei Duan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Guangli Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xiaoyan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Linyu Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Hao Zheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Chongyang Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Jiaolin Wen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Ning Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Cheng Peng
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Xia Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- * E-mail:
| |
Collapse
|
47
|
Mathematical modeling to distinguish cell cycle arrest and cell killing in chemotherapeutic concentration response curves. J Pharmacokinet Pharmacodyn 2011; 38:385-403. [DOI: 10.1007/s10928-011-9199-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Accepted: 04/16/2011] [Indexed: 12/18/2022]
|
48
|
Toward a clinically feasible gene expression-based subclassification strategy for septic shock: proof of concept. Crit Care Med 2010; 38:1955-61. [PMID: 20639748 DOI: 10.1097/ccm.0b013e3181eb924f] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To develop a clinically feasible stratification strategy for pediatric septic shock, using gene expression mosaics and a 100-gene signature representing the first 24 hrs of admission to the pediatric intensive care unit. DESIGN Prospective, observational study involving microarray-based bioinformatics. SETTING Multiple pediatric intensive care units in the United States. PATIENTS Ninety-eight children with septic shock. INTERVENTIONS None other than standard care. MEASUREMENTS AND MAIN RESULTS Patients were classified into three previously published, genome-wide, expression-based subclasses (subclasses A, B, and C) having clinically relevant phenotypic differences. The class-defining 100-gene signature was depicted for each individual patient, using mosaics generated by the Gene Expression Dynamics Inspector (GEDI). Composite mosaics were generated representing the average expression patterns for each of the three subclasses. Nine individual clinicians served as blinded evaluators. Each evaluator was shown the 98 individual patient mosaics and asked to classify each patient into one of the three subclasses, using the composite mosaics as the reference point. The respective sensitivities, specificities, positive predictive values, and negative predictive values of the subclassification strategy were ≥ 4% across the three subclasses. The classification strategy also generated positive likelihood ratios of ≥ 6.8 and negative likelihood ratios of ≤ .2 across the three subclasses. The κ coefficient across all possible interevaluator comparisons was 0.81. CONCLUSIONS We have provided initial evidence (proof of concept) for a clinically feasible and robust stratification strategy for pediatric septic shock based on a 100-gene signature and gene expression mosaics.
Collapse
|
49
|
Ning T, Yan X, Lu ZJ, Wang GP, Zhang NG, Yang JL, Jiang SS, Wu Y, Yang L, Guan YS, Luo F. Gene therapy with the angiogenesis inhibitor endostatin in an orthotopic lung cancer murine model. Hum Gene Ther 2010; 20:103-11. [PMID: 18939902 DOI: 10.1089/hum.2008.098] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Angiogenesis plays an important role in the growth of solid tumors. To date, no information has been acquired on the effectiveness of gene therapy in the orthotopic lung cancer model of syngeneic immunocompetent mice treated with an angiogenesis inhibitor. Here, we report the establishment of such a model in which Lewis lung carcinoma (LL/2) cell suspensions were orthotopically inoculated into the lung parenchyma of C57BL/6 mice, which were also injected with a recombinant adenoviral vector delivering the human endostatin gene (Ad-hE). We found that orthotopic implantation of LL/2 cells into the lung parenchyma produced a solitary tumor nodule in the lung followed by remote mediastinal lymph node metastasis. Conditioned medium from Ad-hE-transfected LL/2 cells apparently inhibited proliferation of human umbilical vein endothelial cells (HUVECs). The level of endostatin protein in serum could be identified by enzyme-linked immunosorbent assay. Treatment with Ad-hE resulted in inhibition of tumor growth and prolongation of survival time of tumor-bearing mice. Immunohistochemical analysis revealed that intratumoral angiogenesis was significantly suppressed. Furthermore, the finding of angiogenesis inhibition was also supported by measuring the number of circulating endothelial cells (CECs). Apoptotic cells were found to be increased within tumor tissues from mice treated with Ad-hE. In addition, treatment with Ad-hE combined with cis-diamminedichloroplatinum(II) (cisplatin) enhanced antitumor activity. These observations provide further evidence of the antitumor effect of endostatin gene therapy, and may be of importance for further exploration of potential application of this combined approach in the treatment of human lung cancer as well as other solid tumors.
Collapse
Affiliation(s)
- Tao Ning
- Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
PROVENCIO M, SANCHEZ A, GARRIDO P, VALCARCEL F, 丁 燕, 李 军. 新型分子靶向药物联合放疗在肺癌中的应用. CHINESE JOURNAL OF LUNG CANCER 2010; 13. [PMCID: PMC6135956 DOI: 10.3779/j.issn.1009-3419.2010.08.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
非小细胞肺癌(non-small cell lung cancer, NSCLC)约占肺癌总病例数的80%-85%,对于Ⅲ期患者来说,NSCLC约占肺癌总病例数的40%。不可切除Ⅲ期NSCLC的治疗为以铂类为基础的化疗联合胸部放疗。本文将综述正在研发中且有可能用于联合治疗的新型靶向制剂。其中最具前景的策略之一为表皮生长因子受体(epidermal growth factor receptor, EGFR)通路的抑制。放疗可激活EGFR信号,通过诱导细胞增殖并增强DNA修复而导致放疗抵抗。几项临床前模型研究表明西妥昔单抗与放疗联合具有协同效应。几项Ⅱ期试验评估了西妥昔单抗与放疗同步使用的安全性与疗效,结果喜人。吉非替尼对多种细胞系具有放疗增敏作用,其与放疗的联合已被试验用于不可切除Ⅲ期NSCLC的治疗。然而,放化疗后使用吉非替尼作为维持治疗的结果不容乐观。一项Ⅰ期试验评估了厄洛替尼与放化疗联合的疗效。放疗可通过损伤细胞膜、DNA以及微血管内皮细胞而诱导肿瘤死亡,而这反过来可增加促血管生成生长因子的产生。抗血管生长制剂可降低血管密度,但可改善肿瘤的含氧量。应用血管内皮生长因子受体(vascular endothelial growth factor receptor, VEGFR)抑制剂可通过阻断亚致死量辐射损伤的修复而增强放疗对人NSCLC的疗效。厄洛替尼、贝伐珠单抗与胸部放疗联合试验正在进行中。该三种药物联合治疗的新策略尚需制订。由于放疗可增强HSP90分子伴侣的功能从而引起肺癌细胞的放疗抵抗,此通路的阻断剂可通过抑制HIF-1α和VEGF的表达进而抑制肺癌细胞的生存和血管生成,因而可能用于减少放疗抵抗。在NSCLC和间皮瘤的临床前模型中,Aurora激酶抑制剂似乎对放疗具有增效作用。
Collapse
Affiliation(s)
- Mariano PROVENCIO
- Department of Medical Oncology, Hospital Universitario Clínica Puerta de Hierro-Majadahonda,Mariano PROVENCIO, MD, PhD, Servicio de Oncología Médica, Hospital Universitario Puerta de Hierro-Majadahonda, Calle Manuel de Falla, 1, Madrid 28222, Spain, Fax: 34-91-344-5190, E-mail:
| | - Antonio SANCHEZ
- Department of Medical Oncology, Hospital Universitario Clínica Puerta de Hierro-Majadahonda
| | | | - Francisco VALCARCEL
- Department of Radiation Oncology, Hospital Universitario Clínica Puerta de Hierro-Majadahonda Madrid, Spain
| | - 燕 丁
- 天津医科大学总医院,天津市肺癌研究所,天津市肺癌转移与肿瘤微环境重点实验室
| | | |
Collapse
|