1
|
Zhan T, Song W, Jing G, Yuan Y, Kang N, Zhang Q. Zebrafish live imaging: a strong weapon in anticancer drug discovery and development. Clin Transl Oncol 2024; 26:1807-1835. [PMID: 38514602 DOI: 10.1007/s12094-024-03406-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/07/2024] [Indexed: 03/23/2024]
Abstract
Developing anticancer drugs is a complex and time-consuming process. The inability of current laboratory models to reflect important aspects of the tumor in vivo limits anticancer medication research. Zebrafish is a rapid, semi-automated in vivo screening platform that enables the use of non-invasive imaging methods to monitor morphology, survival, developmental status, response to drugs, locomotion, or other behaviors. Zebrafish models are widely used in drug discovery and development for anticancer drugs, especially in conjunction with live imaging techniques. Herein, we concentrated on the use of zebrafish live imaging in anticancer therapeutic research, including drug screening, efficacy assessment, toxicity assessment, and mechanism studies. Zebrafish live imaging techniques have been used in numerous studies, but this is the first time that these techniques have been comprehensively summarized and compared side by side. Finally, we discuss the hypothesis of Zebrafish Composite Model, which may provide future directions for zebrafish imaging in the field of cancer research.
Collapse
Affiliation(s)
- Tiancheng Zhan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Wanqian Song
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Guo Jing
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Yongkang Yuan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China
| | - Ning Kang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China.
| | - Qiang Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China.
| |
Collapse
|
2
|
Yvone GM, Breunig JJ. Pediatric low-grade glioma models: advances and ongoing challenges. Front Oncol 2024; 13:1346949. [PMID: 38318325 PMCID: PMC10839015 DOI: 10.3389/fonc.2023.1346949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/29/2023] [Indexed: 02/07/2024] Open
Abstract
Pediatric low-grade gliomas represent the most common childhood brain tumor class. While often curable, some tumors fail to respond and even successful treatments can have life-long side effects. Many clinical trials are underway for pediatric low-grade gliomas. However, these trials are expensive and challenging to organize due to the heterogeneity of patients and subtypes. Advances in sequencing technologies are helping to mitigate this by revealing the molecular landscapes of mutations in pediatric low-grade glioma. Functionalizing these mutations in the form of preclinical models is the next step in both understanding the disease mechanisms as well as for testing therapeutics. However, such models are often more difficult to generate due to their less proliferative nature, and the heterogeneity of tumor microenvironments, cell(s)-of-origin, and genetic alterations. In this review, we discuss the molecular and genetic alterations and the various preclinical models generated for the different types of pediatric low-grade gliomas. We examined the different preclinical models for pediatric low-grade gliomas, summarizing the scientific advances made to the field and therapeutic implications. We also discuss the advantages and limitations of the various models. This review highlights the importance of preclinical models for pediatric low-grade gliomas while noting the challenges and future directions of these models to improve therapeutic outcomes of pediatric low-grade gliomas.
Collapse
Affiliation(s)
- Griselda Metta Yvone
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Joshua J. Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
3
|
Dudziak K, Nowak M, Sozoniuk M. One Host-Multiple Applications: Zebrafish ( Danio rerio) as Promising Model for Studying Human Cancers and Pathogenic Diseases. Int J Mol Sci 2022; 23:10255. [PMID: 36142160 PMCID: PMC9499349 DOI: 10.3390/ijms231810255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/03/2022] [Accepted: 09/03/2022] [Indexed: 11/17/2022] Open
Abstract
In recent years, zebrafish (ZF) has been increasingly applied as a model in human disease studies, with a particular focus on cancer. A number of advantages make it an attractive alternative for mice widely used so far. Due to the many advantages of zebrafish, modifications can be based on different mechanisms and the induction of human disease can take different forms depending on the research goal. Genetic manipulation, tumor transplantation, or injection of the pathogen are only a few examples of using ZF as a model. Most of the studies are conducted in order to understand the disease mechanism, monitor disease progression, test new or alternative therapies, and select the best treatment. The transplantation of cancer cells derived from patients enables the development of personalized medicine. To better mimic a patient's body environment, immune-deficient models (SCID) have been developed. A lower immune response is mostly generated by genetic manipulation but also by irradiation or dexamethasone treatment. For many studies, using SCID provides a better chance to avoid cancer cell rejection. In this review, we describe the main directions of using ZF in research, explain why and how zebrafish can be used as a model, what kind of limitations will be met and how to overcome them. We collected recent achievements in this field, indicating promising perspectives for the future.
Collapse
Affiliation(s)
- Karolina Dudziak
- Chair and Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Michał Nowak
- Institute of Plant Genetics, Breeding and Biotechnology, University of Life Sciences in Lublin, 20-950 Lublin, Poland
| | - Magdalena Sozoniuk
- Institute of Plant Genetics, Breeding and Biotechnology, University of Life Sciences in Lublin, 20-950 Lublin, Poland
| |
Collapse
|
4
|
Basheer F, Dhar P, Samarasinghe RM. Zebrafish Models of Paediatric Brain Tumours. Int J Mol Sci 2022; 23:9920. [PMID: 36077320 PMCID: PMC9456103 DOI: 10.3390/ijms23179920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
Paediatric brain cancer is the second most common childhood cancer and is the leading cause of cancer-related deaths in children. Despite significant advancements in the treatment modalities and improvements in the 5-year survival rate, it leaves long-term therapy-associated side effects in paediatric patients. Addressing these impairments demands further understanding of the molecularity and heterogeneity of these brain tumours, which can be demonstrated using different animal models of paediatric brain cancer. Here we review the use of zebrafish as potential in vivo models for paediatric brain tumour modelling, as well as catalogue the currently available zebrafish models used to study paediatric brain cancer pathophysiology, and discuss key findings, the unique attributes that these models add, current challenges and therapeutic significance.
Collapse
Affiliation(s)
- Faiza Basheer
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Instiute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3220, Australia
| | - Poshmaal Dhar
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Instiute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3220, Australia
| | - Rasika M. Samarasinghe
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Instiute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3220, Australia
| |
Collapse
|
5
|
Precision Medicine in Head and Neck Cancers: Genomic and Preclinical Approaches. J Pers Med 2022; 12:jpm12060854. [PMID: 35743639 PMCID: PMC9224778 DOI: 10.3390/jpm12060854] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/11/2022] [Accepted: 05/19/2022] [Indexed: 02/07/2023] Open
Abstract
Head and neck cancers (HNCs) represent the sixth most widespread malignancy worldwide. Surgery, radiotherapy, chemotherapeutic and immunotherapeutic drugs represent the main clinical approaches for HNC patients. Moreover, HNCs are characterised by an elevated mutational load; however, specific genetic mutations or biomarkers have not yet been found. In this scenario, personalised medicine is showing its efficacy. To study the reliability and the effects of personalised treatments, preclinical research can take advantage of next-generation sequencing and innovative technologies that have been developed to obtain genomic and multi-omic profiles to drive personalised treatments. The crosstalk between malignant and healthy components, as well as interactions with extracellular matrices, are important features which are responsible for treatment failure. Preclinical research has constantly implemented in vitro and in vivo models to mimic the natural tumour microenvironment. Among them, 3D systems have been developed to reproduce the tumour mass architecture, such as biomimetic scaffolds and organoids. In addition, in vivo models have been changed over the last decades to overcome problems such as animal management complexity and time-consuming experiments. In this review, we will explore the new approaches aimed to improve preclinical tools to study and apply precision medicine as a therapeutic option for patients affected by HNCs.
Collapse
|
6
|
Larsson S, Kettunen P, Carén H. Orthotopic Transplantation of Human Paediatric High-Grade Glioma in Zebrafish Larvae. Brain Sci 2022; 12:brainsci12050625. [PMID: 35625011 PMCID: PMC9139401 DOI: 10.3390/brainsci12050625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/30/2022] [Accepted: 05/07/2022] [Indexed: 02/01/2023] Open
Abstract
Brain tumours are the most common cause of death among children with solid tumours, and high-grade gliomas (HGG) are among the most devastating forms with very poor outcomes. In the search for more effective treatments for paediatric HGG, there is a need for better experimental models. To date, there are no xenograft zebrafish models developed for human paediatric HGG; existing models rely on adult cells. The use of paediatric models is of great importance since it is well known that the genetic and epigenetic mechanisms behind adult and paediatric disease differ greatly. In this study, we present a clinically relevant in vivo model based on paediatric primary glioma stem cell (GSC) cultures, which after orthotopic injection into the zebrafish larvae, can be monitored using confocal imaging over time. We show that cells invade the brain tissue and can be followed up to 8 days post-injection while they establish in the fore/mid brain. This model offers an in vivo system where tumour invasion can be monitored and drug treatments quickly be evaluated. The possibility to monitor patient-specific cells has the potential to contribute to a better understanding of cellular behaviour and personalised treatments in the future.
Collapse
Affiliation(s)
- Susanna Larsson
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden;
| | - Petronella Kettunen
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
- Department of Neuropathology, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Helena Carén
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden;
- Correspondence: ; Tel.: +46-31-786-3838
| |
Collapse
|
7
|
Mendonça-Gomes JM, Valverde TM, Martins TMDM, Charlie-Silva I, Padovani BN, Fénero CM, da Silva EM, Domingues RZ, Melo-Hoyos DC, Corrêa-Junior JD, Câmara NOS, Góes AM, Gomes DA. Long-term dexamethasone treatment increases the engraftment efficiency of human breast cancer cells in adult zebrafish. FISH AND SHELLFISH IMMUNOLOGY REPORTS 2021; 2:100007. [DOI: 10.1016/j.fsirep.2021.100007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/03/2021] [Accepted: 03/03/2021] [Indexed: 01/03/2023] Open
|
8
|
Shahzad U, Taccone MS, Kumar SA, Okura H, Krumholtz S, Ishida J, Mine C, Gouveia K, Edgar J, Smith C, Hayes M, Huang X, Derry WB, Taylor MD, Rutka JT. Modeling human brain tumors in flies, worms, and zebrafish: From proof of principle to novel therapeutic targets. Neuro Oncol 2021; 23:718-731. [PMID: 33378446 DOI: 10.1093/neuonc/noaa306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
For decades, cell biologists and cancer researchers have taken advantage of non-murine species to increase our understanding of the molecular processes that drive normal cell and tissue development, and when perturbed, cause cancer. The advent of whole-genome sequencing has revealed the high genetic homology of these organisms to humans. Seminal studies in non-murine organisms such as Drosophila melanogaster, Caenorhabditis elegans, and Danio rerio identified many of the signaling pathways involved in cancer. Studies in these organisms offer distinct advantages over mammalian cell or murine systems. Compared to murine models, these three species have shorter lifespans, are less resource intense, and are amenable to high-throughput drug and RNA interference screening to test a myriad of promising drugs against novel targets. In this review, we introduce species-specific breeding strategies, highlight the advantages of modeling brain tumors in each non-mammalian species, and underscore the successes attributed to scientific investigation using these models. We conclude with an optimistic proposal that discoveries in the fields of cancer research, and in particular neuro-oncology, may be expedited using these powerful screening tools and strategies.
Collapse
Affiliation(s)
- Uswa Shahzad
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada.,Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Michael S Taccone
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Sachin A Kumar
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Hidehiro Okura
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Stacey Krumholtz
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Joji Ishida
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Coco Mine
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Kyle Gouveia
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Julia Edgar
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Christian Smith
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Madeline Hayes
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - Xi Huang
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada.,Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - W Brent Derry
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Michael D Taylor
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - James T Rutka
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada.,Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| |
Collapse
|
9
|
Yuan M, White D, Resar L, Bar E, Groves M, Cohen A, Jackson E, Bynum J, Rubens J, Mumm J, Chen L, Jiang L, Raabe E, Rodriguez FJ, Eberhart CG. Conditional reprogramming culture conditions facilitate growth of lower-grade glioma models. Neuro Oncol 2021; 23:770-782. [PMID: 33258947 DOI: 10.1093/neuonc/noaa263] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The conditional reprogramming cell culture method was developed to facilitate growth of senescence-prone normal and neoplastic epithelial cells, and involves co-culture with irradiated fibroblasts and the addition of a small molecule Rho kinase (ROCK) inhibitor. The aim of this study was to determine whether this approach would facilitate the culture of compact low-grade gliomas. METHODS We attempted to culture 4 pilocytic astrocytomas, 2 gangliogliomas, 2 myxopapillary ependymomas, 2 anaplastic gliomas, 2 difficult-to-classify low-grade neuroepithelial tumors, a desmoplastic infantile ganglioglioma, and an anaplastic pleomorphic xanthoastrocytoma using a modified conditional reprogramming cell culture approach. RESULTS Conditional reprogramming resulted in robust increases in growth for a majority of these tumors, with fibroblast conditioned media and ROCK inhibition both required. Switching cultures to standard serum containing media, or serum-free neurosphere conditions, with or without ROCK inhibition, resulted in decreased proliferation and induction of senescence markers. Rho kinase inhibition and conditioned media both promoted Akt and Erk1/2 activation. Several cultures, including one derived from a NF1-associated pilocytic astrocytoma (JHH-NF1-PA1) and one from a BRAF p.V600E mutant anaplastic pleomorphic xanthoastrocytoma (JHH-PXA1), exhibited growth sufficient for preclinical testing in vitro. In addition, JHH-NF1-PA1 cells survived and migrated in larval zebrafish orthotopic xenografts, while JHH-PXA1 formed orthotopic xenografts in mice histopathologically similar to the tumor from which it was derived. CONCLUSIONS These studies highlight the potential for the conditional reprogramming cell culture method to promote the growth of glial and glioneuronal tumors in vitro, in some cases enabling the establishment of long-term culture and in vivo models.
Collapse
Affiliation(s)
- Ming Yuan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David White
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Linda Resar
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eli Bar
- Department of Pathology, University of Maryland, Baltimore, Maryland, USA
| | - Mari Groves
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alan Cohen
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eric Jackson
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jennifer Bynum
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeffrey Rubens
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeff Mumm
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Liam Chen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Liqun Jiang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eric Raabe
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Fausto J Rodriguez
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Li Z, Langhans SA. In Vivo and Ex Vivo Pediatric Brain Tumor Models: An Overview. Front Oncol 2021; 11:620831. [PMID: 33869004 PMCID: PMC8047472 DOI: 10.3389/fonc.2021.620831] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 03/15/2021] [Indexed: 12/18/2022] Open
Abstract
After leukemia, tumors of the brain and spine are the second most common form of cancer in children. Despite advances in treatment, brain tumors remain a leading cause of death in pediatric cancer patients and survivors often suffer from life-long consequences of side effects of therapy. The 5-year survival rates, however, vary widely by tumor type, ranging from over 90% in more benign tumors to as low as 20% in the most aggressive forms such as glioblastoma. Even within historically defined tumor types such as medulloblastoma, molecular analysis identified biologically heterogeneous subgroups each with different genetic alterations, age of onset and prognosis. Besides molecularly driven patient stratification to tailor disease risk to therapy intensity, such a diversity demonstrates the need for more precise and disease-relevant pediatric brain cancer models for research and drug development. Here we give an overview of currently available in vitro and in vivo pediatric brain tumor models and discuss the opportunities that new technologies such as 3D cultures and organoids that can bridge limitations posed by the simplicity of monolayer cultures and the complexity of in vivo models, bring to accommodate better precision in drug development for pediatric brain tumors.
Collapse
Affiliation(s)
| | - Sigrid A. Langhans
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| |
Collapse
|
11
|
Tucker ER, George S, Angelini P, Bruna A, Chesler L. The Promise of Patient-Derived Preclinical Models to Accelerate the Implementation of Personalised Medicine for Children with Neuroblastoma. J Pers Med 2021; 11:248. [PMID: 33808071 PMCID: PMC8065808 DOI: 10.3390/jpm11040248] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 01/02/2023] Open
Abstract
Patient-derived preclinical models are now a core component of cancer research and have the ability to drastically improve the predictive power of preclinical therapeutic studies. However, their development and maintenance can be challenging, time consuming, and expensive. For neuroblastoma, a developmental malignancy of the neural crest, it is possible to establish patient-derived models as xenografts in mice and zebrafish, and as spheroids and organoids in vitro. These varied approaches have contributed to comprehensive packages of preclinical evidence in support of new therapeutics for neuroblastoma. We discuss here the ethical and technical considerations for the creation of patient-derived models of neuroblastoma and how their use can be optimized for the study of tumour evolution and preclinical therapies. We also discuss how neuroblastoma patient-derived models might become avatars for personalised medicine for children with this devastating disease.
Collapse
Affiliation(s)
- Elizabeth R. Tucker
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, Cotswold Road, London SM2 5NG, UK; (E.R.T.); (S.G.)
| | - Sally George
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, Cotswold Road, London SM2 5NG, UK; (E.R.T.); (S.G.)
| | - Paola Angelini
- Children and Young People’s Unit, The Royal Marsden, Downs Road, Sutton, Surrey SM2 5PT, UK;
| | - Alejandra Bruna
- Preclinical Paediatric Cancer Evolution, Centre for Cancer Drug Discovery, The Institute of Cancer Research, Cotswold Road, London SM2 5NG, UK;
| | - Louis Chesler
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, Cotswold Road, London SM2 5NG, UK; (E.R.T.); (S.G.)
| |
Collapse
|
12
|
Chen Z, Peng P, Zhang X, Mania-Farnell B, Xi G, Wan F. Advanced Pediatric Diffuse Pontine Glioma Murine Models Pave the Way towards Precision Medicine. Cancers (Basel) 2021; 13:cancers13051114. [PMID: 33807733 PMCID: PMC7961799 DOI: 10.3390/cancers13051114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) account for ~15% of pediatric brain tumors, which invariably present with poor survival regardless of treatment mode. Several seminal studies have revealed that 80% of DIPGs harbor H3K27M mutation coded by HIST1H3B, HIST1H3C and H3F3A genes. The H3K27M mutation has broad effects on gene expression and is considered a tumor driver. Determination of the effects of H3K27M on posttranslational histone modifications and gene regulations in DIPG is critical for identifying effective therapeutic targets. Advanced animal models play critical roles in translating these cutting-edge findings into clinical trial development. Here, we review current molecular research progress associated with DIPG. We also summarize DIPG animal models, highlighting novel genomic engineered mouse models (GEMMs) and innovative humanized DIPG mouse models. These models will pave the way towards personalized precision medicine for the treatment of DIPGs.
Collapse
Affiliation(s)
- Zirong Chen
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
| | - Peng Peng
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
| | - Xiaolin Zhang
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
| | - Barbara Mania-Farnell
- Department of Biological Science, Purdue University Northwest, Hammond, IN 46323, USA;
| | - Guifa Xi
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Correspondence: (G.X.); (F.W.); Tel.: +1-(312)5034296 (G.X.); +86-(027)-8366-5201 (F.W.)
| | - Feng Wan
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
- Correspondence: (G.X.); (F.W.); Tel.: +1-(312)5034296 (G.X.); +86-(027)-8366-5201 (F.W.)
| |
Collapse
|
13
|
Reimunde P, Pensado-López A, Carreira Crende M, Lombao Iglesias V, Sánchez L, Torrecilla-Parra M, Ramírez CM, Anfray C, Torres Andón F. Cellular and Molecular Mechanisms Underlying Glioblastoma and Zebrafish Models for the Discovery of New Treatments. Cancers (Basel) 2021; 13:1087. [PMID: 33802571 PMCID: PMC7961726 DOI: 10.3390/cancers13051087] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is the most common of all brain malignant tumors; it displays a median survival of 14.6 months with current complete standard treatment. High heterogeneity, aggressive and invasive behavior, the impossibility of completing tumor resection, limitations for drug administration and therapeutic resistance to current treatments are the main problems presented by this pathology. In recent years, our knowledge of GBM physiopathology has advanced significantly, generating relevant information on the cellular heterogeneity of GBM tumors, including cancer and immune cells such as macrophages/microglia, genetic, epigenetic and metabolic alterations, comprising changes in miRNA expression. In this scenario, the zebrafish has arisen as a promising animal model to progress further due to its unique characteristics, such as transparency, ease of genetic manipulation, ethical and economic advantages and also conservation of the major brain regions and blood-brain-barrier (BBB) which are similar to a human structure. A few papers described in this review, using genetic and xenotransplantation zebrafish models have been used to study GBM as well as to test the anti-tumoral efficacy of new drugs, their ability to interact with target cells, modulate the tumor microenvironment, cross the BBB and/or their toxicity. Prospective studies following these lines of research may lead to a better diagnosis, prognosis and treatment of patients with GBM.
Collapse
Affiliation(s)
- Pedro Reimunde
- Department of Medicine, Campus de Oza, Universidade da Coruña, 15006 A Coruña, Spain
- Department of Neurosurgery, Hospital Universitario Lucus Augusti, 27003 Lugo, Spain
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Martín Carreira Crende
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Vanesa Lombao Iglesias
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Marta Torrecilla-Parra
- IMDEA Research Institute of Food and Health Sciences, 28049 Madrid, Spain; (M.T.-P.); (C.M.R.)
| | - Cristina M. Ramírez
- IMDEA Research Institute of Food and Health Sciences, 28049 Madrid, Spain; (M.T.-P.); (C.M.R.)
| | - Clément Anfray
- IRCCS Istituto Clinico Humanitas, Via A. Manzoni 56, 20089 Rozzano, Milan, Italy;
| | - Fernando Torres Andón
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
- IRCCS Istituto Clinico Humanitas, Via A. Manzoni 56, 20089 Rozzano, Milan, Italy;
| |
Collapse
|
14
|
Chen X, Li Y, Yao T, Jia R. Benefits of Zebrafish Xenograft Models in Cancer Research. Front Cell Dev Biol 2021; 9:616551. [PMID: 33644052 PMCID: PMC7905065 DOI: 10.3389/fcell.2021.616551] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
As a promising in vivo tool for cancer research, zebrafish have been widely applied in various tumor studies. The zebrafish xenograft model is a low-cost, high-throughput tool for cancer research that can be established quickly and requires only a small sample size, which makes it favorite among researchers. Zebrafish patient-derived xenograft (zPDX) models provide promising evidence for short-term clinical treatment. In this review, we discuss the characteristics and advantages of zebrafish, such as their transparent and translucent features, the use of vascular fluorescence imaging, the establishment of metastatic and intracranial orthotopic models, individual pharmacokinetics measurements, and tumor microenvironment. Furthermore, we introduce how these characteristics and advantages are applied other in tumor studies. Finally, we discuss the future direction of the use of zebrafish in tumor studies and provide new ideas for the application of it.
Collapse
Affiliation(s)
- Xingyu Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yongyun Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Tengteng Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
15
|
Studying the Tumor Microenvironment in Zebrafish. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:69-92. [PMID: 34664234 DOI: 10.1007/978-3-030-73119-9_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The tumor microenvironment significantly contributes to tumor initiation, progression, neo-angiogenesis, and metastasis, and a better understanding of the role of the different cellular players would facilitate the development of novel therapeutic strategies for cancer treatment. Towards this goal, intravital imaging is a powerful method to unravel interaction partners of tumor cells. Among vertebrate model organisms, zebrafish is uniquely suited for in vivo imaging studies. In recent years zebrafish has also become a valuable model in cancer research. In this chapter, we will summarize, how zebrafish has been used to characterize cells of the tumor microenvironment. We will cover both genetically engineered cancer models and xenograft models in zebrafish. The majority of work has been done on the role of innate immune cells and their role during tumor initiation and metastasis, but we will also cover studies focusing on adipocytes, fibroblasts, and endothelial cells. Taken together, we will highlight the versatile use of the zebrafish model for in vivo tumor microenvironment studies.
Collapse
|
16
|
Rapid In Vivo Validation of HDAC Inhibitor-Based Treatments in Neuroblastoma Zebrafish Xenografts. Pharmaceuticals (Basel) 2020; 13:ph13110345. [PMID: 33121173 PMCID: PMC7692187 DOI: 10.3390/ph13110345] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 01/01/2023] Open
Abstract
The survival rate among children with relapsed neuroblastomas continues to be poor, and thus new therapeutic approaches identified by reliable preclinical drug testing models are urgently needed. Zebrafish are a powerful vertebrate model in preclinical cancer research. Here, we describe a zebrafish neuroblastoma yolk sac model to evaluate efficacy and toxicity of histone deacetylase (HDAC) inhibitor treatments. Larvae were engrafted with fluorescently labeled, genetically diverse, established cell lines and short-term cultures of patient-derived primary cells. Engrafted tumors progressed locally and disseminated remotely in an intact environment. Combination treatments involving the standard chemotherapy doxorubicin and HDAC inhibitors substantially reduced tumor volume, induced tumor cell death, and inhibited tumor cell dissemination to the tail region. Hence, this model allows for fast, cost-efficient, and reliable in vivo evaluation of toxicity and response of the primary and metastatic tumor sites to drug combinations.
Collapse
|
17
|
Targen S, Kaya T, Avci ME, Gunes D, Keskus AG, Konu O. ZenoFishDb v1.1: A Database for Xenotransplantation Studies in Zebrafish. Zebrafish 2020; 17:305-318. [PMID: 32931381 DOI: 10.1089/zeb.2020.1869] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Rapidly accumulating literature has proven feasibility of the zebrafish xenograft models in cancer research. Nevertheless, online databases for searching the current zebrafish xenograft literature are in great demand. Herein, we have developed a manually curated database, called ZenoFishDb v1.1 (https://konulab.shinyapps.io/zenofishdb), based on R Shiny platform aiming to provide searchable information on ever increasing collection of zebrafish studies for cancer cell line transplantation and patient-derived xenografts (PDXs). ZenoFishDb v1.1 user interface contains four modules: DataTable, Visualization, PDX Details, and PDX Charts. The DataTable and Visualization pages represent xenograft study details, including injected cell lines, PDX injections, molecular modifications of cell lines, zebrafish strains, as well as technical aspects of the xenotransplantation procedures in table, bar, and/or pie chart formats. The PDX Details module provides comprehensive information on the patient details in table format and can be searched and visualized. Overall, ZenoFishDb v1.1 enables researchers to effectively search, list, and visualize different technical and biological attributes of zebrafish xenotransplantation studies particularly focusing on the new trends that make use of reporters, RNA interference, overexpression, or mutant gene constructs of transplanted cancer cells, stem cells, and PDXs, as well as distinguished host modifications.
Collapse
Affiliation(s)
- Seniye Targen
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Tuğberk Kaya
- Interdisciplinary Program in Neuroscience, Bilkent University, Ankara, Turkey.,Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - M Ender Avci
- Izmir Biomedicine and Genome Center, Dokuz Eylul University, Izmir, Turkey
| | - Damla Gunes
- Interdisciplinary Program in Neuroscience, Bilkent University, Ankara, Turkey
| | - Ayse Gokce Keskus
- Interdisciplinary Program in Neuroscience, Bilkent University, Ankara, Turkey
| | - Ozlen Konu
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey.,Interdisciplinary Program in Neuroscience, Bilkent University, Ankara, Turkey.,UNAM-Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| |
Collapse
|
18
|
Cabezas-Sáinz P, Pensado-López A, Sáinz B, Sánchez L. Modeling Cancer Using Zebrafish Xenografts: Drawbacks for Mimicking the Human Microenvironment. Cells 2020; 9:E1978. [PMID: 32867288 PMCID: PMC7564051 DOI: 10.3390/cells9091978] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/07/2020] [Accepted: 08/19/2020] [Indexed: 02/07/2023] Open
Abstract
The first steps towards establishing xenografts in zebrafish embryos were performed by Lee et al., 2005 and Haldi et al., 2006, paving the way for studying human cancers using this animal species. Since then, the xenograft technique has been improved in different ways, ranging from optimizing the best temperature for xenografted embryo incubation, testing different sites for injection of human tumor cells, and even developing tools to study how the host interacts with the injected cells. Nonetheless, a standard protocol for performing xenografts has not been adopted across laboratories, and further research on the temperature, microenvironment of the tumor or the cell-host interactions inside of the embryo during xenografting is still needed. As a consequence, current non-uniform conditions could be affecting experimental results in terms of cell proliferation, invasion, or metastasis; or even overestimating the effects of some chemotherapeutic drugs on xenografted cells. In this review, we highlight and raise awareness regarding the different aspects of xenografting that need to be improved in order to mimic, in a more efficient way, the human tumor microenvironment, resulting in more robust and accurate in vivo results.
Collapse
Affiliation(s)
- Pablo Cabezas-Sáinz
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (A.P.-L.)
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (A.P.-L.)
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Bruno Sáinz
- Departamento de Bioquímica, Facultad de Medicina, Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC-UAM, Universidad Autónoma de Madrid, Arzobispo Morcillo 4, 28029 Madrid, Spain;
- Cancer Stem Cell and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer Area 3-Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (A.P.-L.)
| |
Collapse
|
19
|
Gronseth E, Gupta A, Koceja C, Kumar S, Kutty RG, Rarick K, Wang L, Ramchandran R. Astrocytes influence medulloblastoma phenotypes and CD133 surface expression. PLoS One 2020; 15:e0235852. [PMID: 32628717 PMCID: PMC7337293 DOI: 10.1371/journal.pone.0235852] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/24/2020] [Indexed: 12/28/2022] Open
Abstract
The medulloblastoma (MB) microenvironment is diverse, and cell-cell interactions within this milieu is of prime importance. Astrocytes, a major component of the microenvironment, have been shown to impact primary tumor cell phenotypes and metastasis. Based on proximity of MB cells and astrocytes in the brain microenvironment, we investigated whether astrocytes may influence MB cell phenotypes directly. Astrocyte conditioned media (ACM) increased Daoy MB cell invasion, adhesion, and in vivo cellular protrusion formation. ACM conditioning of MB cells also increased CD133 surface expression, a key cancer stem cell marker of MB. Additional neural stem cell markers, Nestin and Oct-4A, were also increased by ACM conditioning, as well as neurosphere formation. By knocking down CD133 using short interfering RNA (siRNA), we showed that ACM upregulated CD133 expression in MB plays an important role in invasion, adhesion and neurosphere formation. Collectively, our data suggests that astrocytes influence MB cell phenotypes by regulating CD133 expression, a key protein with defined roles in MB tumorgenicity and survival.
Collapse
Affiliation(s)
- Emily Gronseth
- Department of Pediatrics, Division of Neonatology, Developmental Vascular Biology Program, Children’s Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Ankan Gupta
- Department of Pediatrics, Division of Neonatology, Developmental Vascular Biology Program, Children’s Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Chris Koceja
- Versiti, Milwaukee, Wisconsin, United States of America
| | - Suresh Kumar
- Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Raman G. Kutty
- Medical College of Wisconsin Affiliated Hospitals, Acsension St. Joseph Hospital, Milwaukee, Wisconsin, United States of America
| | - Kevin Rarick
- Department of Pediatrics, Division of Critical Care, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Ling Wang
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Ramani Ramchandran
- Department of Pediatrics, Division of Neonatology, Developmental Vascular Biology Program, Children’s Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
20
|
Houston Z, Bunt J, Chen KS, Puttick S, Howard CB, Fletcher NL, Fuchs AV, Cui J, Ju Y, Cowin G, Song X, Boyd AW, Mahler SM, Richards LJ, Caruso F, Thurecht KJ. Understanding the Uptake of Nanomedicines at Different Stages of Brain Cancer Using a Modular Nanocarrier Platform and Precision Bispecific Antibodies. ACS CENTRAL SCIENCE 2020; 6:727-738. [PMID: 32490189 PMCID: PMC7256936 DOI: 10.1021/acscentsci.9b01299] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Indexed: 06/11/2023]
Abstract
Increasing accumulation and retention of nanomedicines within tumor tissue is a significant challenge, particularly in the case of brain tumors where access to the tumor through the vasculature is restricted by the blood-brain barrier (BBB). This makes the application of nanomedicines in neuro-oncology often considered unfeasible, with efficacy limited to regions of significant disease progression and compromised BBB. However, little is understood about how the evolving tumor-brain physiology during disease progression affects the permeability and retention of designer nanomedicines. We report here the development of a modular nanomedicine platform that, when used in conjunction with a unique model of how tumorigenesis affects BBB integrity, allows investigation of how nanomaterial properties affect uptake and retention in brain tissue. By combining different in vivo longitudinal imaging techniques (including positron emission tomography and magnetic resonance imaging), we have evaluated the retention of nanomedicines with predefined physicochemical properties (size and surface functionality) and established a relationship between structure and tissue accumulation as a function of a new parameter that measures BBB leakiness; this offers significant advancements in our ability to relate tumor accumulation of nanomedicines to more physiologically relevant parameters. Our data show that accumulation of nanomedicines in brain tumor tissue is better correlated with the leakiness of the BBB than actual tumor volume. This was evaluated by establishing brain tumors using a spontaneous and endogenously derived glioblastoma model providing a unique opportunity to assess these parameters individually and compare the results across multiple mice. We also quantitatively demonstrate that smaller nanomedicines (20 nm) can indeed cross the BBB and accumulate in tumors at earlier stages of the disease than larger analogues, therefore opening the possibility of developing patient-specific nanoparticle treatment interventions in earlier stages of the disease. Importantly, these results provide a more predictive approach for designing efficacious personalized nanomedicines based on a particular patient's condition.
Collapse
Affiliation(s)
- Zachary
H. Houston
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jens Bunt
- Queensland
Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Kok-Siong Chen
- Queensland
Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
- Brigham
and Women’s Hospital, Harvard Medical
School, Boston, Massachusetts 02115, United States
| | - Simon Puttick
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- Commonwealth
Scientific and Industrial Research Organisation, Probing Biosystems
Future Science Platform, Royal Brisbane
and Women’s Hospital, Brisbane, Queensland 4029, Australia
| | - Christopher B. Howard
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC Training
Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC Training Centre for Biopharmaceutical
Innovation The University
of Queensland, St Lucia, Queensland 4072, Australia
| | - Nicholas L. Fletcher
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Adrian V. Fuchs
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jiwei Cui
- Department
of Chemical Engineering, The University
of Melbourne, Parkville, Victoria 3010, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
- Key
Laboratory of Colloid and Interface Chemistry of the Ministry of Education,
School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Yi Ju
- Department
of Chemical Engineering, The University
of Melbourne, Parkville, Victoria 3010, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Gary Cowin
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
| | - Xin Song
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
| | - Andrew W. Boyd
- Leukaemia
Foundation Laboratory, QIMR-Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
- Department
of Medicine, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Stephen M. Mahler
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC Training Centre for Biopharmaceutical
Innovation The University
of Queensland, St Lucia, Queensland 4072, Australia
| | - Linda J. Richards
- Queensland
Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
- The
School of Biomedical Sciences, The University
of Queensland, St Lucia, Queensland 4072, Australia
| | - Frank Caruso
- Department
of Chemical Engineering, The University
of Melbourne, Parkville, Victoria 3010, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Kristofer J. Thurecht
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC Training
Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
21
|
Fan JJ, Hsu WH, Lee KH, Chen KC, Lin CW, Lee YLA, Ko TP, Lee LT, Lee MT, Chang MS, Cheng CH. Dietary Flavonoids Luteolin and Quercetin Inhibit Migration and Invasion of Squamous Carcinoma through Reduction of Src/Stat3/S100A7 Signaling. Antioxidants (Basel) 2019; 8:antiox8110557. [PMID: 31731716 PMCID: PMC6912538 DOI: 10.3390/antiox8110557] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/13/2019] [Accepted: 11/13/2019] [Indexed: 12/29/2022] Open
Abstract
Flavonoids are well-known antioxidants and have shown the ability to prevent tumor formation and recurrence. Especially in dietary flavonoids, they have provided convenience and consistence of intake for long-term prevention of tumor formation. Previous reports suggested that S100 calcium-binding protein A7 (S100A7) might activate epithelial–mesenchymal transition (EMT) signaling and promote the metastasis of tumor cells; however, the regulatory signaling was unclear. In this study, we found that S100A7 was highly expressed in cancer cells and could be reduced by luteolin (Lu) and quercetin (Qu) through Src/Stat3 signaling. We found that the protein levels of S100A7, phosphorylated Src (p-Src), and p-Stat3 were increased in A431-III cells. Flavonoids Lu and Qu reduce protein levels of p-Src, p-Stat3 and S100A7 in A431-III cells. Treatment of A431-III cells with Src inhibitor SU6656 and Stat3 inhibitor S3I-201 also reduced the protein levels of S100A7. Transactivation activity of 5′-upstream regions of S100A7 was activated by Stat3 but was reduced by treatment with Lu, Qu, SU6656 and S3I-201. The treatment also reduced the migratory and invasive abilities of A431-III cells. In a further analysis of EMT markers, the protein level of E-cad increased and that of Twist decreased after treatment with the inhibitors and flavonoids. Overexpression of S100A7 decreased the protein level of E-cad and increased the Twist level, whereas knockdown of S100A7 had the opposite effects. Treatment with S3I-201, Lu and Qu, compared to the control, were found to decrease metastasis of tumor cells in zebrafish larvae. These results suggest that Lu and Qu may inhibit Src/Stat3/S100A7 signaling to reduce tumorigenesis of cancer cells.
Collapse
Affiliation(s)
- Jhen-Jia Fan
- Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan;
- Food and Drug Administration, Ministry of Health and Welfare, Taipei 11561, Taiwan
| | - Wen-Hsien Hsu
- Department of Surgery, Wan-Fang Hospital, Taipei Medical University, Taipei 11034, Taiwan;
| | - Kuen-Haur Lee
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11034, Taiwan;
- Cancer Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 11034, Taiwan
| | - Ku-Chung Chen
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11034, Taiwan; (K.-C.C.); (C.-W.L.)
| | - Cheng-Wei Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11034, Taiwan; (K.-C.C.); (C.-W.L.)
| | - Yu-Lin A Lee
- Departments of Medicine and Pediatrics, Duke University Hospital, Durham, NC 27704, USA;
| | - Tzu-Ping Ko
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; (T.-P.K.); (M.-T.L.)
| | - Lang-Ta Lee
- Department of Nursing, Ching Kuo Institute of Management and Health, Keelung 20301, Taiwan;
| | - Ming-Ting Lee
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; (T.-P.K.); (M.-T.L.)
| | - Mau-Sun Chang
- Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan;
- Correspondence: (C.-H.C.); (M.-S.C.); Tel.: +886-2-27361661 (ext. 3156) (C.-H.C.); +886-2-33669837 (M.-S.C.)
| | - Chia-Hsiung Cheng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11034, Taiwan; (K.-C.C.); (C.-W.L.)
- Correspondence: (C.-H.C.); (M.-S.C.); Tel.: +886-2-27361661 (ext. 3156) (C.-H.C.); +886-2-33669837 (M.-S.C.)
| |
Collapse
|
22
|
Pudelko L, Edwards S, Balan M, Nyqvist D, Al-Saadi J, Dittmer J, Almlöf I, Helleday T, Bräutigam L. An orthotopic glioblastoma animal model suitable for high-throughput screenings. Neuro Oncol 2019; 20:1475-1484. [PMID: 29750281 DOI: 10.1093/neuonc/noy071] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Glioblastoma (GBM) is an aggressive form of brain cancer with poor prognosis. Although murine animal models have given valuable insights into the GBM disease biology, they cannot be used in high-throughput screens to identify and profile novel therapies. The only vertebrate model suitable for large-scale screens, the zebrafish, has proven to faithfully recapitulate biology and pathology of human malignancies, and clinically relevant orthotopic zebrafish models have been developed. However, currently available GBM orthotopic zebrafish models do not support high-throughput drug discovery screens. Methods We transplanted both GBM cell lines as well as patient-derived material into zebrafish blastulas. We followed the behavior of the transplants with time-lapse microscopy and real-time in vivo light-sheet microscopy. Results We found that GBM material transplanted into zebrafish blastomeres robustly migrated into the developing nervous system, establishing an orthotopic intracranial tumor already 24 hours after transplantation. Detailed analysis revealed that our model faithfully recapitulates the human disease. Conclusion We have developed a robust, fast, and automatable transplantation assay to establish orthotopic GBM tumors in zebrafish. In contrast to currently available orthotopic zebrafish models, our approach does not require technically challenging intracranial transplantation of single embryos. Our improved zebrafish model enables transplantation of thousands of embryos per hour, thus providing an orthotopic vertebrate GBM model for direct application in drug discovery screens.
Collapse
Affiliation(s)
- Linda Pudelko
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Steven Edwards
- Department of Applied Physics, Science for Life Laboratory, Royal Institute of Technology, Stockholm, Sweden
| | - Mirela Balan
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Daniel Nyqvist
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jonathan Al-Saadi
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Johannes Dittmer
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid Almlöf
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Helleday
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Lars Bräutigam
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Letrado P, de Miguel I, Lamberto I, Díez-Martínez R, Oyarzabal J. Zebrafish: Speeding Up the Cancer Drug Discovery Process. Cancer Res 2018; 78:6048-6058. [PMID: 30327381 DOI: 10.1158/0008-5472.can-18-1029] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/29/2018] [Accepted: 08/23/2018] [Indexed: 11/16/2022]
Abstract
Zebrafish (Danio rerio) is an ideal in vivo model to study a wide variety of human cancer types. In this review, we provide a comprehensive overview of zebrafish in the cancer drug discovery process, from (i) approaches to induce malignant tumors, (ii) techniques to monitor cancer progression, and (iii) strategies for compound administration to (iv) a compilation of the 355 existing case studies showing the impact of zebrafish models on cancer drug discovery, which cover a broad scope of scenarios. Finally, based on the current state-of-the-art analysis, this review presents some highlights about future directions using zebrafish in cancer drug discovery and the potential of this model as a prognostic tool in prospective clinical studies. Cancer Res; 78(21); 6048-58. ©2018 AACR.
Collapse
Affiliation(s)
- Patricia Letrado
- Ikan Biotech SL, The Zebrafish Lab Department, Centro Europeo de Empresas e Innovación de Navarra (CEIN), Noain, Spain.,Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Irene de Miguel
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Iranzu Lamberto
- Ikan Biotech SL, The Zebrafish Lab Department, Centro Europeo de Empresas e Innovación de Navarra (CEIN), Noain, Spain
| | - Roberto Díez-Martínez
- Ikan Biotech SL, The Zebrafish Lab Department, Centro Europeo de Empresas e Innovación de Navarra (CEIN), Noain, Spain.
| | - Julen Oyarzabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.
| |
Collapse
|
24
|
Dobson THW, Gopalakrishnan V. Preclinical Models of Pediatric Brain Tumors-Forging Ahead. Bioengineering (Basel) 2018; 5:E81. [PMID: 30279402 PMCID: PMC6315787 DOI: 10.3390/bioengineering5040081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/22/2018] [Accepted: 09/27/2018] [Indexed: 12/11/2022] Open
Abstract
Approximately five out of 100,000 children from 0 to 19 years old are diagnosed with a brain tumor. These children are treated with medication designed for adults that are highly toxic to a developing brain. Those that survive are at high risk for a lifetime of limited physical, psychological, and cognitive abilities. Despite much effort, not one drug exists that was designed specifically for pediatric patients. Stagnant government funding and the lack of economic incentives for the pharmaceutical industry greatly limits preclinical research and the development of clinically applicable pediatric brain tumor models. As more data are collected, the recognition of disease sub-groups based on molecular heterogeneity increases the need for designing specific models suitable for predictive drug screening. To overcome these challenges, preclinical approaches will need continual enhancement. In this review, we examine the advantages and shortcomings of in vitro and in vivo preclinical pediatric brain tumor models and explore potential solutions based on past, present, and future strategies for improving their clinical relevancy.
Collapse
Affiliation(s)
- Tara H W Dobson
- Department of Pediatrics, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Vidya Gopalakrishnan
- Department of Pediatrics, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
- Department of Molecular & Cellular Oncology, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
- Brain Tumor Center, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
- Center for Cancer Epigenetics, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
- Graduate School of Biomedical Sciences UT-Health Science Center, Houston, TX 77030, USA.
| |
Collapse
|
25
|
Chia K, Mazzolini J, Mione M, Sieger D. Tumor initiating cells induce Cxcr4-mediated infiltration of pro-tumoral macrophages into the brain. eLife 2018; 7:e31918. [PMID: 29465400 PMCID: PMC5821457 DOI: 10.7554/elife.31918] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/31/2018] [Indexed: 12/28/2022] Open
Abstract
It is now clear that microglia and macrophages are present in brain tumors, but whether or how they affect initiation and development of tumors is not known. Exploiting the advantages of the zebrafish (Danio rerio) model, we showed that macrophages and microglia respond immediately upon oncogene activation in the brain. Overexpression of human AKT1 within neural cells of larval zebrafish led to a significant increase in the macrophage and microglia populations. By using a combination of transgenic and mutant zebrafish lines, we showed that this increase was caused by the infiltration of peripheral macrophages into the brain mediated via Sdf1b-Cxcr4b signaling. Intriguingly, confocal live imaging reveals highly dynamic interactions between macrophages/microglia and pre-neoplastic cells, which do not result in phagocytosis of pre-neoplastic cells. Finally, depletion of macrophages and microglia resulted in a significant reduction of oncogenic cell proliferation. Thus, macrophages and microglia show tumor promoting functions already during the earliest stages of the developing tumor microenvironment.
Collapse
Affiliation(s)
- Kelda Chia
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Julie Mazzolini
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Marina Mione
- Centre for Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Dirk Sieger
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
26
|
Cagan R, Meyer P. Rethinking cancer: current challenges and opportunities in cancer research. Dis Model Mech 2017; 10:349-352. [PMID: 28381596 PMCID: PMC5399574 DOI: 10.1242/dmm.030007] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Cancer therapeutics currently have the lowest clinical trial success rate of all major diseases. Partly as a result of the paucity of successful anti-cancer drugs, cancer will soon be the leading cause of mortality in developed countries. As a disease embedded in the fundamentals of our biology, cancer presents difficult challenges that would benefit from uniting experts from a broad cross-section of related and unrelated fields. Combining extant approaches with novel ones could help in tackling this challenging health problem, enabling the development of therapeutics to stop disease progression and prolong patient lives. This goal provided the inspiration for a recent workshop titled ‘Rethinking Cancer’, which brought together a group of cancer scientists who work in the academic and pharmaceutical sectors of Europe, America and Asia. In this Editorial, we discuss the main themes emerging from the workshop, with the aim of providing a snapshot of key challenges faced by the cancer research community today. We also outline potential strategies for addressing some of these challenges, from understanding the basic evolution of cancer and improving its early detection to streamlining the thorny process of moving promising drug targets into clinical trials. Summary: Pablo Meyer and Ross Cagan discuss the main themes emerging from a recent workshop ‘Rethinking Cancer’, highlighting the key challenges faced by the research community and outlining potential strategies to promote translation of basic findings.
Collapse
Affiliation(s)
- Ross Cagan
- Icahn School of Medicine at Mount Sinai, Annenberg 25-40, Campus Box 1020, 1468 Madison Avenue, New York, NY 10029, USA
| | - Pablo Meyer
- Icahn School of Medicine at Mount Sinai, Annenberg 25-40, Campus Box 1020, 1468 Madison Avenue, New York, NY 10029, USA .,Thomas J. Watson Computational Biology Center, IBM, Yorktown Heights, NY 10598, USA
| |
Collapse
|
27
|
Astone M, Dankert EN, Alam SK, Hoeppner LH. Fishing for cures: The alLURE of using zebrafish to develop precision oncology therapies. NPJ Precis Oncol 2017; 1. [PMID: 29376139 PMCID: PMC5784449 DOI: 10.1038/s41698-017-0043-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Zebrafish have proven to be a valuable model to study human cancer biology with the ultimate aim of developing new therapies. Danio rerio are amenable to in vivo imaging, high-throughput drug screening, mutagenesis, and transgenesis, and they share histological and genetic similarities with Homo sapiens. The significance of zebrafish in the field of precision oncology is rapidly emerging. Indeed, modeling cancer in zebrafish has already been used to identify tumor biomarkers, define therapeutic targets and provide an in vivo platform for drug discovery. New zebrafish studies are starting to pave the way to direct individualized clinical applications. Patient-derived cancer cell xenograft models have demonstrated the feasibility of using zebrafish as a real-time avatar of prognosis and drug response to identify the most ideal therapy for an individual patient. Genetic cancer modeling in zebrafish, now facilitated by rapidly evolving genome editing techniques, represents another innovative approach to recapitulate human oncogenesis and develop individualized treatments. Utilizing zebrafish to design customizable precision therapies will improve the clinical outcome of patients afflicted with cancer.
Collapse
Affiliation(s)
- Matteo Astone
- The Hormel Institute, University of Minnesota, Austin, MN, 55912
| | - Erin N Dankert
- The Hormel Institute, University of Minnesota, Austin, MN, 55912
| | - Sk Kayum Alam
- The Hormel Institute, University of Minnesota, Austin, MN, 55912
| | - Luke H Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN, 55912
| |
Collapse
|
28
|
Miserocchi G, Mercatali L, Liverani C, De Vita A, Spadazzi C, Pieri F, Bongiovanni A, Recine F, Amadori D, Ibrahim T. Management and potentialities of primary cancer cultures in preclinical and translational studies. J Transl Med 2017; 15:229. [PMID: 29116016 PMCID: PMC5688825 DOI: 10.1186/s12967-017-1328-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/27/2017] [Indexed: 02/07/2023] Open
Abstract
The use of patient-derived primary cell cultures in cancer preclinical assays has increased in recent years. The management of resected tumor tissue remains complex and a number of parameters must be respected to obtain complete sample digestion and optimal vitality yield. We provide an overview of the benefits of correct primary cell culture management using different preclinical methodologies, and describe the pros and cons of this model with respect to other kinds of samples. One important advantage is that the heterogeneity of the cell populations composing a primary culture partially reproduces the tumor microenvironment and crosstalk between malignant and healthy cells, neither of which is possible with cell lines. Moreover, the use of patient-derived specimens in innovative preclinical technologies, such as 3D systems or bioreactors, represents an important opportunity to improve the translational value of the results obtained. In vivo models could further our understanding of the crosstalk between tumor and other tissues as they enable us to observe the systemic and biological interactions of a complete organism. Although engineered mice are the most common model used in this setting, the zebrafish (Danio rerio) species has recently been recognized as an innovative experimental system. In fact, the transparent body and incomplete immune system of zebrafish embryos are especially useful for evaluating patient-derived tumor tissue interactions in healthy hosts. In conclusion, ex vivo systems represent an important tool for cancer research, but samples require correct manipulation to maximize their translational value.
Collapse
Affiliation(s)
- Giacomo Miserocchi
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Laura Mercatali
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy.
| | - Chiara Liverani
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Alessandro De Vita
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Chiara Spadazzi
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Federica Pieri
- Pathology Unit, Morgagni-Pierantoni Hospital, Via Carlo Forlanini 34, 47121, Forlì, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Federica Recine
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Dino Amadori
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| |
Collapse
|
29
|
Wenger A, Larsson S, Danielsson A, Elbæk KJ, Kettunen P, Tisell M, Sabel M, Lannering B, Nordborg C, Schepke E, Carén H. Stem cell cultures derived from pediatric brain tumors accurately model the originating tumors. Oncotarget 2017; 8:18626-18639. [PMID: 28148893 PMCID: PMC5386635 DOI: 10.18632/oncotarget.14826] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 01/16/2017] [Indexed: 12/14/2022] Open
Abstract
Brain tumors are the leading cause of cancer-related death in children but high-grade gliomas in children and adolescents have remained a relatively under-investigated disease despite this. A better understanding of the cellular and molecular pathogenesis of the diseases is required in order to improve the outcome for these children. In vitro-cultured primary tumor cells from patients are indispensable tools for this purpose by enabling functional analyses and development of new therapies. However, relevant well-characterized in vitro cultures from pediatric gliomas cultured under serum-free conditions have been lacking. We have therefore established patient-derived in vitro cultures and performed thorough characterization of the cells using large-scale analyses of DNA methylation, copy-number alterations and investigated their stability during prolonged time in culture. We show that the cells were stable during prolonged culture in serum-free stem cell media without apparent alterations in morphology or growth rate. The cells were proliferative, positive for stem cell markers, able to respond to differentiation cues and initiated tumors in zebrafish and mice suggesting that the cells are cancer stem cells or progenitor cells. The cells accurately mirrored the tumor they were derived from in terms of methylation pattern, copy number alterations and DNA mutations. These unique primary in vitro cultures can thus be used as a relevant and robust model system for functional studies on pediatric brain tumors.
Collapse
Affiliation(s)
- Anna Wenger
- Department of Pathology, Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Susanna Larsson
- Department of Pathology, Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Danielsson
- Department of Oncology, Sahlgrenska Cancer Center, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kirstine Juul Elbæk
- Department of Pathology, Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Petronella Kettunen
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Neuropathology, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Magnus Tisell
- Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magnus Sabel
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,The Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Birgitta Lannering
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,The Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Claes Nordborg
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Elizabeth Schepke
- Department of Pathology, Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,The Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Helena Carén
- Department of Pathology, Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
30
|
Benjamin DC, Hynes RO. Intravital imaging of metastasis in adult Zebrafish. BMC Cancer 2017; 17:660. [PMID: 28946867 PMCID: PMC5613480 DOI: 10.1186/s12885-017-3647-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 09/13/2017] [Indexed: 11/25/2022] Open
Abstract
Background Metastasis is a major clinical problem whose biology is not yet fully understood. This lack of understanding is especially true for the events at the metastatic site, which include arrest, extravasation, and growth into macrometastases. Intravital imaging is a powerful technique that has shown great promise in increasing our understanding of these events. To date, most intravital imaging studies have been performed in mice, which has limited its adoption. Zebrafish are also a common system for the intravital imaging of metastasis. However, as imaging in embryos is technically simpler, relatively few studies have used adult zebrafish to study metastasis and none have followed individual cells at the metastatic site over time. The aim of this study was to demonstrate that adult casper zebrafish offer a convenient model system for performing intravital imaging of the metastatic site over time with single-cell resolution. Methods ZMEL1 zebrafish melanoma cells were injected into 6 to 10-week-old casper fish using an intravenous injection protocol. Because casper fish are transparent even as adults, they could be imaged without surgical intervention. Individual cells were followed over the course of 2 weeks as they arrested, extravasated, and formed macroscopic metastases. Results Our injection method reliably delivered cells into circulation and led to the formation of tumors in multiple organs. Cells in the skin and sub-dermal muscle could be imaged at high resolution over 2 weeks using confocal microscopy. Arrest was visualized and determined to be primarily due to size restriction. Following arrest, extravasation was seen to occur between 1 and 6 days post-injection. Once outside of the vasculature, cells were observed migrating as well as forming protrusions. Conclusions Casper fish are a useful model for studying the events at the metastatic site using intravital imaging. The protocols described in this study are relatively simple. Combined with the reasonably low cost of zebrafish, they offer to increase access to intravital imaging. Electronic supplementary material The online version of this article (10.1186/s12885-017-3647-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David C Benjamin
- Department of Biology, Massachusetts Institute of Technology, 31 Ames Street, Cambridge, MA, 02139, USA.,David H. Koch Institute For Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA.,Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, MD, 20815, USA
| | - Richard O Hynes
- Department of Biology, Massachusetts Institute of Technology, 31 Ames Street, Cambridge, MA, 02139, USA. .,David H. Koch Institute For Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA. .,Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
31
|
Zebrafish in Translational Cancer Research: Insight into Leukemia, Melanoma, Glioma and Endocrine Tumor Biology. Genes (Basel) 2017; 8:genes8090236. [PMID: 28930163 PMCID: PMC5615369 DOI: 10.3390/genes8090236] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 09/08/2017] [Accepted: 09/14/2017] [Indexed: 02/06/2023] Open
Abstract
Over the past 15 years, zebrafish have emerged as a powerful tool for studying human cancers. Transgenic techniques have been employed to model different types of tumors, including leukemia, melanoma, glioblastoma and endocrine tumors. These models present histopathological and molecular conservation with their human cancer counterparts and have been fundamental for understanding mechanisms of tumor initiation and progression. Moreover, xenotransplantation of human cancer cells in embryos or adult zebrafish offers the advantage of studying the behavior of human cancer cells in a live organism. Chemical-genetic screens using zebrafish embryos have uncovered novel druggable pathways and new therapeutic strategies, some of which are now tested in clinical trials. In this review, we will report on recent advances in using zebrafish as a model in cancer studies—with specific focus on four cancer types—where zebrafish has contributed to novel discoveries or approaches to novel therapies.
Collapse
|
32
|
Casey MJ, Modzelewska K, Anderson D, Goodman J, Boer EF, Jimenez L, Grossman D, Stewart RA. Transplantation of Zebrafish Pediatric Brain Tumors into Immune-competent Hosts for Long-term Study of Tumor Cell Behavior and Drug Response. J Vis Exp 2017. [PMID: 28570545 PMCID: PMC5607995 DOI: 10.3791/55712] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Tumor cell transplantation is an important technique to define the mechanisms controlling cancer cell growth, migration, and host response, as well as to assess potential patient response to therapy. Current methods largely depend on using syngeneic or immune-compromised animals to avoid rejection of the tumor graft. Such methods require the use of specific genetic strains that often prevent the analysis of immune-tumor cell interactions and/or are limited to specific genetic backgrounds. An alternative method in zebrafish takes advantage of an incompletely developed immune system in the embryonic brain before 3 days, where tumor cells are transplanted for use in short-term assays (i.e., 3 to 10 days). However, these methods cause host lethality, which prevents the long-term study of tumor cell behavior and drug response. This protocol describes a simple and efficient method for the long-term orthotopic transplantation of zebrafish brain tumor tissue into the fourth ventricle of a 2-day-old immune-competent zebrafish. This method allows: 1) long-term study of tumor cell behaviors, such as invasion and dissemination; 2) durable tumor response to drugs; and 3) re-transplantation of tumors for the study of tumor evolution and/or the impact of different host genetic backgrounds. In summary, this technique allows cancer researchers to assess engraftment, invasion, and growth at distant sites, as well as to perform chemical screens and cell competition assays over many months. This protocol can be extended to studies of other tumor types and can be used to elucidate mechanisms of chemoresistance and metastasis.
Collapse
Affiliation(s)
- Mattie J Casey
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine
| | - Katarzyna Modzelewska
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine
| | - Daniela Anderson
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine
| | - James Goodman
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine
| | - Elena F Boer
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine
| | - Laura Jimenez
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine
| | - Douglas Grossman
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine; Department of Dermatology, University of Utah Health Sciences Center, Salt Lake City
| | - Rodney A Stewart
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine;
| |
Collapse
|
33
|
Lenting K, Verhaak R, Ter Laan M, Wesseling P, Leenders W. Glioma: experimental models and reality. Acta Neuropathol 2017; 133:263-282. [PMID: 28074274 PMCID: PMC5250671 DOI: 10.1007/s00401-017-1671-4] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/06/2017] [Accepted: 01/06/2017] [Indexed: 12/12/2022]
Abstract
In theory, in vitro and in vivo models for human gliomas have great potential to not only enhance our understanding of glioma biology, but also to facilitate the development of novel treatment strategies for these tumors. For reliable prediction and validation of the effects of different therapeutic modalities, however, glioma models need to comply with specific and more strict demands than other models of cancer, and these demands are directly related to the combination of genetic aberrations and the specific brain micro-environment gliomas grow in. This review starts with a brief introduction on the pathological and molecular characteristics of gliomas, followed by an overview of the models that have been used in the last decades in glioma research. Next, we will discuss how these models may play a role in better understanding glioma development and especially in how they can aid in the design and optimization of novel therapies. The strengths and weaknesses of the different models will be discussed in light of genotypic, phenotypic and metabolic characteristics of human gliomas. The last part of this review provides some examples of how therapy experiments using glioma models can lead to deceptive results when such characteristics are not properly taken into account.
Collapse
Affiliation(s)
- Krissie Lenting
- Department of Pathology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Roel Verhaak
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Mark Ter Laan
- Department of Neurosurgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Pathology, Princess Máxima Center for Pediatric Oncology and University Medical Center Utrecht, Utrecht, The Netherlands
| | - William Leenders
- Department of Pathology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
34
|
Chen L, Groenewoud A, Tulotta C, Zoni E, Kruithof-de Julio M, van der Horst G, van der Pluijm G, Ewa Snaar-Jagalska B. A zebrafish xenograft model for studying human cancer stem cells in distant metastasis and therapy response. Methods Cell Biol 2016; 138:471-496. [PMID: 28129855 DOI: 10.1016/bs.mcb.2016.10.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lethal and incurable bone metastasis is one of the main causes of death in multiple types of cancer. A small subpopulation of cancer stem/progenitor-like cells (CSCs), also known as tumor-initiating cells from heterogenetic cancer is considered to mediate bone metastasis. Although over the past decades numerous studies have been performed in different types of cancer, it is still difficult to track small numbers of CSCs during the onset of metastasis. With use of noninvasive high-resolution imaging, transparent zebrafish embryos can be employed to dynamically visualize cancer progression and reciprocal interaction with stroma in a living organism. Recently we established a zebrafish CSC-xenograft model to visually and functionally analyze the role of CSCs and their interactions with the microenvironment at the onset of metastasis. Given the highly conserved human and zebrafish genome, transplanted human cancer cells are able to respond to zebrafish cytokines, modulate the zebrafish microenvironment, and take advantage of the zebrafish stroma during cancer progression. This chapter delineates the zebrafish CSC-xenograft model as a useful tool for both CSC biological study and anticancer drug screening.
Collapse
Affiliation(s)
- L Chen
- Leiden University, Leiden, The Netherlands
| | | | - C Tulotta
- Leiden University, Leiden, The Netherlands
| | - E Zoni
- University of Bern, Bern, Switzerland; Leiden University Medical Centre, Leiden, The Netherlands
| | - M Kruithof-de Julio
- University of Bern, Bern, Switzerland; Leiden University Medical Centre, Leiden, The Netherlands
| | | | | | | |
Collapse
|
35
|
Hwang SJ, Park HG, Park Y, Lee HJ. An α-quaternary chiral latam derivative, YH-304 as a novel broad-spectrum anticancer agent. Int J Oncol 2016; 49:2480-2486. [PMID: 27748805 DOI: 10.3892/ijo.2016.3726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/30/2016] [Indexed: 11/06/2022] Open
Abstract
Previously, we reported that α-quaternary chiral lactam derivatives have broad spectrum anticancer activity. However, the underlying molecular mechanisms and its relevance are largely unknown. In the present study, we report progress on α-quaternary chiral lactam analogues that address this, focusing on the novel analogue YH-304 as a candidate to broadly target human cancer cells. The effect of YH-304 on cell transformation was assessed by clonogenic assay in non-small cell lung cancer cells (NSCLCs) A549 and 226B. Proapoptotic activity of YH-304 was determined by TUNEL assay and cleaved PARP, cleaved caspase-9, and Bax as markers for apoptosis. The p53-dependency and therapeutic spectrum of YH-304 was assessed by western blot analysis, real-time PCR, and cell viability assays in cells expressing endogenous wild or mutant p53. The effect of YH-304 on angiogenesis in vivo was examined by bFGF-mediated angiogenesis assay in zebrafish. Finally, the effect of YH-304 on AKT and ERK activation (phosphorylation) as a putative mechanism underlying the effect of YH-304 on bFGF-mediated angiogenesis was assessed using western blotting. We found that YH-304 significantly decreases the colony-forming activities of both A549 and 226B cells, inducing cellular apoptosis. Unlike nutlin-3 (p53 pathway activator), YH-304 did not affect the expression levels of p53 and its target gene such as p21 and thus showed p53-independent anticancer activity with broad spectrum. In addition, YH-304 inhibited bFGF-induced angiogenesis in vivo through mediating AKT and ERK signaling pathway, which plays an important role in bFGF activation and angiogenesis. Taken together, our data indicate that YH-304 may represent a novel therapeutic option for the treatment of cancer in a p53-independent manner.
Collapse
Affiliation(s)
- Su Jung Hwang
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam 621-749, Republic of Korea
| | - Hyeung-Geun Park
- Research Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Yohan Park
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam 621-749, Republic of Korea
| | - Hyo-Jong Lee
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam 621-749, Republic of Korea
| |
Collapse
|
36
|
Tulotta C, He S, van der Ent W, Chen L, Groenewoud A, Spaink HP, Snaar-Jagalska BE. Imaging Cancer Angiogenesis and Metastasis in a Zebrafish Embryo Model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:239-63. [PMID: 27165357 DOI: 10.1007/978-3-319-30654-4_11] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tumor angiogenesis and metastasis are key steps of cancer progression. In vitro and animal model studies have contributed to partially elucidating the mechanisms involved in these processes and in developing therapies. Besides the improvements in fundamental research and the optimization of therapeutic regimes, cancer still remains a major health threatening condition and therefore the development of new models is needed. The zebrafish is a powerful tool to study tumor angiogenesis and metastasis, because it allows the visualization of fluorescently labelled tumor cells inducing vessel remodeling, disseminating and invading surrounding tissues in a whole transparent embryo. The embryo model has also been used to address the contribution of the tumor stroma in sustaining tumor angiogenesis and spreading. Simultaneously, new anti-angiogenic drugs and compounds affecting malignant cell survival and migration can be tested by simply adding the compound into the water of living embryos. Therefore the zebrafish model offers the opportunity to gain more knowledge on cancer angiogenesis and metastasis in vivo with the final aim of providing new translational insights into therapeutic approaches to help patients.
Collapse
Affiliation(s)
- C Tulotta
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - S He
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - W van der Ent
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - L Chen
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - A Groenewoud
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - H P Spaink
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - B E Snaar-Jagalska
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
37
|
Vittori M, Breznik B, Gredar T, Hrovat K, Bizjak Mali L, Lah TT. Imaging of human glioblastoma cells and their interactions with mesenchymal stem cells in the zebrafish (Danio rerio) embryonic brain. Radiol Oncol 2016; 50:159-67. [PMID: 27247548 PMCID: PMC4852964 DOI: 10.1515/raon-2016-0017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/07/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND An attractive approach in the study of human cancers is the use of transparent zebrafish (Danio rerio) embryos, which enable the visualization of cancer progression in a living animal. MATERIALS AND METHODS We implanted mixtures of fluorescently labeled glioblastoma (GBM) cells and bonemarrow-derived mesenchymal stem cells (MSCs) into zebrafish embryos to study the cellular pathways of their invasion and the interactions between these cells in vivo. RESULTS By developing and applying a carbocyanine-dye-compatible clearing protocol for observation of cells in deep tissues, we showed that U87 and U373 GBM cells rapidly aggregated into tumor masses in the ventricles and midbrain hemispheres of the zebrafish embryo brain, and invaded the central nervous system, often using the ventricular system and the central canal of the spinal cord. However, the GBM cells did not leave the central nervous system. With co-injection of differentially labeled cultured GBM cells and MSCs, the implanted cells formed mixed tumor masses in the brain. We observed tight associations between GBM cells and MSCs, and possible cell-fusion events. GBM cells and MSCs used similar invasion routes in the central nervous system. CONCLUSIONS This simple model can be used to study the molecular pathways of cellular processes in GBM cell invasion, and their interactions with various types of stromal cells in double or triple cell co-cultures, to design anti-GBM cell therapies that use MSCs as vectors.
Collapse
Affiliation(s)
- Milos Vittori
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | | | - Tajda Gredar
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | | | - Lilijana Bizjak Mali
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | | |
Collapse
|
38
|
Stable multilineage xenogeneic replacement of definitive hematopoiesis in adult zebrafish. Sci Rep 2016; 6:19634. [PMID: 26777855 PMCID: PMC4726038 DOI: 10.1038/srep19634] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/16/2015] [Indexed: 11/30/2022] Open
Abstract
Bony fishes are the most numerous and phenotypically diverse group of vertebrates inhabiting our planet, making them an ideal target for identifying general principles of tissue development and function. However, lack of suitable experimental platforms prevents the exploitation of this rich source of natural phenotypic variation. Here, we use a zebrafish strain lacking definitive hematopoiesis for interspecific analysis of hematopoietic cell development. Without conditioning prior to transplantation, hematopoietic progenitor cells from goldfish stably engraft in adult zebrafish homozygous for the c-mybI181N mutation. However, in competitive repopulation experiments, zebrafish hematopoietic cells exhibit an advantage over their goldfish counterparts, possibly owing to subtle species-specific functional differences in hematopoietic microenvironments resulting from over 100 million years of independent evolution. Thus, our unique animal model provides an unprecedented opportunity to genetically and functionally disentangle universal and species-specific contributions of the microenvironment to hematopoietic progenitor cell maintenance and development.
Collapse
|
39
|
Welker AM, Jaros BD, Puduvalli VK, Imitola J, Kaur B, Beattie CE. Standardized orthotopic xenografts in zebrafish reveal glioma cell-line-specific characteristics and tumor cell heterogeneity. Dis Model Mech 2015; 9:199-210. [PMID: 26659251 PMCID: PMC4770147 DOI: 10.1242/dmm.022921] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 12/03/2015] [Indexed: 01/19/2023] Open
Abstract
Glioblastoma (GBM) is a deadly brain cancer, for which few effective drug treatments are available. Several studies have used zebrafish models to study GBM, but a standardized approach to modeling GBM in zebrafish was lacking to date, preventing comparison of data across studies. Here, we describe a new, standardized orthotopic xenotransplant model of GBM in zebrafish. Dose-response survival assays were used to define the optimal number of cells for tumor formation. Techniques to measure tumor burden and cell spread within the brain over real time were optimized using mouse neural stem cells as control transplants. Applying this standardized approach, we transplanted two patient-derived GBM cell lines, serum-grown adherent cells and neurospheres, into the midbrain region of embryonic zebrafish and analyzed transplanted larvae over time. Progressive brain tumor growth and premature larval death were observed using both cell lines; however, fewer transplanted neurosphere cells were needed for tumor growth and lethality. Tumors were heterogeneous, containing both cells expressing stem cell markers and cells expressing markers of differentiation. A small proportion of transplanted neurosphere cells expressed glial fibrillary acidic protein (GFAP) or vimentin, markers of more differentiated cells, but this number increased significantly during tumor growth, indicating that these cells undergo differentiation in vivo. By contrast, most serum-grown adherent cells expressed GFAP and vimentin at the earliest times examined post-transplant. Both cell types produced brain tumors that contained Sox2+ cells, indicative of tumor stem cells. Transplanted larvae were treated with currently used GBM therapeutics, temozolomide or bortezomib, and this resulted in a reduction in tumor volume in vivo and an increase in survival. The standardized model reported here facilitates robust and reproducible analysis of glioblastoma tumor cells in real time and provides a platform for drug screening. Summary: This zebrafish xenotransplant model of glioblastoma enables in vivo imaging of tumor cells and rapid screening for anti-glioma agents. It provides standardization of a model that is easily replicated across laboratories.
Collapse
Affiliation(s)
- Alessandra M Welker
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Brian D Jaros
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Vinay K Puduvalli
- Department of Neurosurgery, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Jaime Imitola
- Department of Neurology, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Balveen Kaur
- Department of Neurosurgery, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Christine E Beattie
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| |
Collapse
|
40
|
Vittori M, Motaln H, Turnšek TL. The study of glioma by xenotransplantation in zebrafish early life stages. J Histochem Cytochem 2015; 63:749-61. [PMID: 26109632 DOI: 10.1369/0022155415595670] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 06/19/2015] [Indexed: 12/31/2022] Open
Abstract
Zebrafish (Danio rerio) and their transparent embryos are becoming an increasingly popular tool for studying processes involved in tumor progression and in the search for novel tumor treatment approaches. The xenotransplantation of fluorescently labeled mammalian cancer cells into zebrafish embryos is an approach enabling relatively high-throughput in vivo analyses. The small size of the embryos as well as the relative simplicity of their manipulation and maintenance allow for large numbers of embryos to be processed efficiently in a short time and at low cost. Furthermore, the possibility of fluorescence microscopic imaging of tumor progression within zebrafish embryos and larvae holds unprecedented potential for the real-time visualization of these processes in vivo. This review presents the methodologies of xenotransplantation studies on zebrafish involving research on tumor invasion, proliferation, tumor-induced angiogenesis and screening for antitumor therapeutics. We further focus on the application of these zebrafish to the study of glioma; in particular, its most common and malignant form, glioblastoma.
Collapse
Affiliation(s)
- Miloš Vittori
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia (MV, HM, TLT)
| | - Helena Motaln
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia (MV, HM, TLT)
| | - Tamara Lah Turnšek
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia (MV, HM, TLT)
| |
Collapse
|
41
|
Barriuso J, Nagaraju R, Hurlstone A. Zebrafish: a new companion for translational research in oncology. Clin Cancer Res 2015; 21:969-75. [PMID: 25573382 DOI: 10.1158/1078-0432.ccr-14-2921] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In an era of high-throughput "omic" technologies, the unprecedented amount of data that can be generated presents a significant opportunity but simultaneously an even greater challenge for oncologists trying to provide personalized treatment. Classically, preclinical testing of new targets and identification of active compounds against those targets have entailed the extensive use of established human cell lines, as well as genetically modified mouse tumor models. Patient-derived xenografts in zebrafish may in the near future provide a platform for selecting an appropriate personalized therapy and together with zebrafish transgenic tumor models represent an alternative vehicle for drug development. The zebrafish is readily genetically modified. The transparency of zebrafish embryos and the recent development of pigment-deficient zebrafish afford researchers the valuable capacity to observe directly cancer formation and progression in a live vertebrate host. The zebrafish is amenable to transplantation assays that test the serial passage of fluorescently labeled tumor cells as well as their capacity to disseminate and/or metastasize. Progress achieved to date in genetic engineering and xenotransplantation will establish the zebrafish as one of the most versatile animal models for cancer research. A model organism that can be used in transgenesis, transplantation assays, single-cell functional assays, and in vivo imaging studies make zebrafish a natural companion for mice in translational oncology research.
Collapse
Affiliation(s)
- Jorge Barriuso
- Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom.
| | - Raghavendar Nagaraju
- Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom
| | - Adam Hurlstone
- Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom.
| |
Collapse
|