1
|
van den Bosch QCC, de Klein A, Verdijk RM, Kiliç E, Brosens E. Uveal melanoma modeling in mice and zebrafish. Biochim Biophys Acta Rev Cancer 2024; 1879:189055. [PMID: 38104908 DOI: 10.1016/j.bbcan.2023.189055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Despite extensive research and refined therapeutic options, the survival for metastasized uveal melanoma (UM) patients has not improved significantly. UM, a malignant tumor originating from melanocytes in the uveal tract, can be asymptomatic and small tumors may be detected only during routine ophthalmic exams; making early detection and treatment difficult. UM is the result of a number of characteristic somatic alterations which are associated with prognosis. Although UM morphology and biology have been extensively studied, there are significant gaps in our understanding of the early stages of UM tumor evolution and effective treatment to prevent metastatic disease remain elusive. A better understanding of the mechanisms that enable UM cells to thrive and successfully metastasize is crucial to improve treatment efficacy and survival rates. For more than forty years, animal models have been used to investigate the biology of UM. This has led to a number of essential mechanisms and pathways involved in UM aetiology. These models have also been used to evaluate the effectiveness of various drugs and treatment protocols. Here, we provide an overview of the molecular mechanisms and pharmacological studies using mouse and zebrafish UM models. Finally, we highlight promising therapeutics and discuss future considerations using UM models such as optimal inoculation sites, use of BAP1mut-cell lines and the rise of zebrafish models.
Collapse
Affiliation(s)
- Quincy C C van den Bosch
- Department of Ophthalmology, Erasmus MC, Rotterdam, the Netherlands; Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands; Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Annelies de Klein
- Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands; Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Robert M Verdijk
- Department of Pathology, Section of Ophthalmic Pathology, Erasmus MC, Rotterdam, The Netherlands; Erasmus MC Cancer Institute, Rotterdam, The Netherlands; Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Emine Kiliç
- Department of Ophthalmology, Erasmus MC, Rotterdam, the Netherlands; Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Erwin Brosens
- Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands; Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Ramos R, Cabré E, Vinyals A, Lorenzo D, Ferreres JR, Varela M, Gomá M, Paules MJ, Gutierrez C, Piulats JM, Fabra À, Caminal JM. Orthotopic murine xenograft model of uveal melanoma with spontaneous liver metastasis. Melanoma Res 2023; 33:1-11. [PMID: 36302215 DOI: 10.1097/cmr.0000000000000860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Uveal melanoma is the most common intraocular malignancy in adults. Despite the effective primary treatment, up to 50% of patients with uveal melanoma will develop metastatic lesions mainly in the liver, which are resistant to conventional chemotherapy and lead to patient's death. To date, no orthotopic murine models of uveal melanoma which can develop spontaneous metastasis are available for preclinical studies. Here, we describe a spontaneous metastatic model of uveal melanoma based on the orthotopic injection of human uveal melanoma cells into the suprachoroidal space of immunodeficient NSG mice. All mice injected with bioluminescent OMM2.5 ( n = 23) or MP41 ( n = 19) cells developed a primary tumor. After eye enucleation, additional bioluminescence signals were detected in the lungs and in the liver. At necropsy, histopathological studies confirmed the presence of lung metastases in 100% of the mice. Liver metastases were assessed in 87 and in 100% of the mice that received OMM2.5 or MP41 cells, respectively. All tumors and metastatic lesions expressed melanoma markers and the signaling molecules insulin-like growth factor type I receptor and myristoylated alanine-rich C-kinase substrate, commonly activated in uveal melanoma. The novelty of this orthotopic mouse xenograft model is the development of spontaneous metastases in the liver from the primary site, reproducing the organoespecificity of metastasis observed in uveal melanoma patients. The faster growth and the high metastatic incidence may be attributed at least in part, to the severe immunodeficiency of NSG mice. This model may be useful for preclinical testing of targeted therapies with potential uveal melanoma antimetastatic activity and to study the mechanisms involved in liver metastasis.
Collapse
Affiliation(s)
- Raquel Ramos
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)
| | - Eduard Cabré
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)
| | - Antònia Vinyals
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)
| | - Daniel Lorenzo
- Ophthalmology Department, Spanish Ocular Oncology National referal center (CSUR) and Ocular Translational Eye Research Unit, Hospital Universitari de Bellvitge (HUB)-IDIBELL
| | | | - Mar Varela
- Pathology Department, Hospital Universitari de Bellvitge
| | - Montse Gomá
- Pathology Department, Hospital Universitari de Bellvitge
| | | | - Cristina Gutierrez
- Radiotherapy Department, Institut Catalá d'Oncologia (ICO), Hospital Duran Reynals
| | - Josep M Piulats
- Medical Oncology, Institut Catalá d'Oncologia (ICO), Hospital Duran Reynals, Barcelona, Spain
| | - Àngels Fabra
- Ophthalmology Department, Spanish Ocular Oncology National referal center (CSUR) and Ocular Translational Eye Research Unit, Hospital Universitari de Bellvitge (HUB)-IDIBELL
| | - José M Caminal
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)
| |
Collapse
|
3
|
Targeting IRS-1/2 in Uveal Melanoma Inhibits In Vitro Cell Growth, Survival and Migration, and In Vivo Tumor Growth. Cancers (Basel) 2022; 14:cancers14246247. [PMID: 36551732 PMCID: PMC9777326 DOI: 10.3390/cancers14246247] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Uveal melanoma originating in the eye and metastasizing to the liver is associated with poor prognosis and has only one approved therapeutic option. We hypothesized that liver-borne growth factors may contribute to UM growth. Therefore, we investigated the role of IGF-1/IGF-1R signaling in UM. Here, we found that IRS-1, the insulin receptor substrate, is overexpressed in both UM cells and tumors. Since we previously observed that IGF-1R antibody therapy was not clinically effective in UM, we investigated the potential of NT157, a small molecule inhibitor of IRS-1/2, in blocking this pathway in UM. NT157 treatment of multiple UM cell lines resulted in reduced cell growth and migration and increased apoptosis. This treatment also significantly inhibited UM tumor growth in vivo, in the chicken egg chorioallantoic membrane (CAM) and subcutaneous mouse models, validating the in vitro effect. Mechanistically, through reverse phase protein array (RPPA), we identified significant proteomic changes in the PI3K/AKT pathway, a downstream mediator of IGF-1 signaling, with NT157 treatment. Together, these results suggest that NT157 inhibits cell growth, survival, and migration in vitro, and tumor growth in vivo via inhibiting IGF-1 signaling in UM.
Collapse
|
4
|
Dewaele S, Delhaye L, De Paepe B, de Bony EJ, De Wilde J, Vanderheyden K, Anckaert J, Yigit N, Nuytens J, Vanden Eynde E, Smet J, Verschoore M, Nemati F, Decaudin D, Rodrigues M, Zhao P, Jochemsen A, Leucci E, Vandesompele J, Van Dorpe J, Marine JC, Van Coster R, Eyckerman S, Mestdagh P. The long non-coding RNA SAMMSON is essential for uveal melanoma cell survival. Oncogene 2022; 41:15-25. [PMID: 34508176 PMCID: PMC8724009 DOI: 10.1038/s41388-021-02006-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 08/02/2021] [Accepted: 08/27/2021] [Indexed: 12/14/2022]
Abstract
Long non-coding RNAs (lncRNAs) can exhibit cell-type and cancer-type specific expression profiles, making them highly attractive as therapeutic targets. Pan-cancer RNA sequencing data revealed broad expression of the SAMMSON lncRNA in uveal melanoma (UM), the most common primary intraocular malignancy in adults. Currently, there are no effective treatments for UM patients with metastatic disease, resulting in a median survival time of 6-12 months. We aimed to investigate the therapeutic potential of SAMMSON inhibition in UM. Antisense oligonucleotide (ASO)-mediated SAMMSON inhibition impaired the growth and viability of a genetically diverse panel of uveal melanoma cell lines. These effects were accompanied by an induction of apoptosis and were recapitulated in two uveal melanoma patient derived xenograft (PDX) models through subcutaneous ASO delivery. SAMMSON pulldown revealed several candidate interaction partners, including various proteins involved in mitochondrial translation. Consequently, inhibition of SAMMSON impaired global, mitochondrial and cytosolic protein translation levels and mitochondrial function in uveal melanoma cells. The present study demonstrates that SAMMSON expression is essential for uveal melanoma cell survival. ASO-mediated silencing of SAMMSON may provide an effective treatment strategy to treat primary and metastatic uveal melanoma patients.
Collapse
Affiliation(s)
- Shanna Dewaele
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Louis Delhaye
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Biotechnology, VIB-Ghent University, Ghent, Belgium
| | - Boel De Paepe
- Department of Pediatrics, Division of Pediatric Neurology and Metabolism, Ghent University Hospital, Ghent, Belgium
| | - Eric James de Bony
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jilke De Wilde
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Department of pathology, Ghent University Hospital, Ghent, Belgium
| | - Katrien Vanderheyden
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jasper Anckaert
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Nurten Yigit
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Justine Nuytens
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Eveline Vanden Eynde
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Joél Smet
- Department of Pediatrics, Division of Pediatric Neurology and Metabolism, Ghent University Hospital, Ghent, Belgium
| | - Maxime Verschoore
- Department of Pediatrics, Division of Pediatric Neurology and Metabolism, Ghent University Hospital, Ghent, Belgium
| | - Fariba Nemati
- Institut Curie, Laboratory of Preclinical Investigation, Translational Research Department, PSL Research University, Paris, France
| | - Didier Decaudin
- Institut Curie, Laboratory of Preclinical Investigation, Translational Research Department, PSL Research University, Paris, France
- Institut Curie, Department of Medical Oncology, PSL Research University, Paris, France
| | - Manuel Rodrigues
- Institut Curie, Department of Medical Oncology, PSL Research University, Paris, France
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, Paris, 75005, France
| | - Peihua Zhao
- Center for Medical Biotechnology, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Aart Jochemsen
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Eleonora Leucci
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
- TRACE, LKI Leuven Cancer Institute, Leuven, Belgium
| | - Jo Vandesompele
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jo Van Dorpe
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of pathology, Ghent University Hospital, Ghent, Belgium
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Department of Oncology, KULeuven, Leuven, Belgium
| | - Rudy Van Coster
- Department of Pediatrics, Division of Pediatric Neurology and Metabolism, Ghent University Hospital, Ghent, Belgium
| | - Sven Eyckerman
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Biotechnology, VIB-Ghent University, Ghent, Belgium
| | - Pieter Mestdagh
- OncoRNALab, Center for Medical Genetics (CMGG), Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.
| |
Collapse
|
5
|
In-vivo imaging for assessing tumor growth in mouse models of ocular melanoma. Exp Eye Res 2021; 204:108431. [PMID: 33406396 DOI: 10.1016/j.exer.2020.108431] [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] [Received: 08/18/2020] [Revised: 11/11/2020] [Accepted: 12/28/2020] [Indexed: 12/14/2022]
Abstract
Uveal melanoma (UM) and conjunctival melanoma (CM) are ocular malignancies that give rise to life-threatening metastases. Although local disease can often be treated successfully, it is often associated with significant vision impairment and treatments are often not effective against metastatic disease. Novel treatment modalities that preserve vision may enable elimination of small tumors and may prevent subsequent metastatic spread. Very few mouse models of metastatic CM and UM are available for research and for development of novel therapies. One of the challenges is to follow tumor growth in-vivo and to determine the right size for treatment, mainly of the posterior, choroidal melanoma. Hence, the purpose of this study was to establish a simple, noninvasive imaging tool that will simplify visualization and tumor follow-up in mouse models of CM and UM. Tumors were induced by inoculation of murine B16LS9 cells into the sub-conjunctival or the choroidal space of a C57BL/6 mouse eye under a surgical microscope. Five to ten days following injection, tumor size was assessed by Phoenix MicronIV™ image-guided Optical Coherence Tomography (OCT) imaging, which included a real-time camera view and OCT scan of the conjunctiva and the retina. In addition, tumor size was evaluated by ultrasound and histopathological examination of eye sections. Tumor growth was observed 5-9 days following sub-conjunctival or sub-retinal injection of seven-thousand or seventy-thousand cells, respectively. A clear tumor mass was detected at these regions using the MicronIV™ imaging system camera and OCT scans. Histology of eye sections confirmed the presence of tumor tissue. OCT allowed an accurate measurement of tumor size in the UM model and a qualitative assessment of tumor size in the CM model. Moreover, OCT enabled assessing the success rate of the choroidal tumor induction and importantly, predicted final tumor size already on the day of cell inoculation. In conclusion, by using a simple, non-invasive imaging tool, we were able to follow intraocular tumor growth of both CM and UM, and to define, already at the time of cell inoculation, a grading scale to evaluate tumor size. This tool may be utilized for evaluation of new mouse models for CM and UM, as well as for testing new therapies for these diseases.
Collapse
|
6
|
Stålhammar G, See TRO, Phillips SS, Grossniklaus HE. Density of PAS positive patterns in uveal melanoma: Correlation with vasculogenic mimicry, gene expression class, BAP-1 expression, macrophage infiltration, and risk for metastasis. Mol Vis 2019; 25:502-516. [PMID: 31588174 PMCID: PMC6776441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/19/2019] [Indexed: 11/22/2022] Open
Abstract
Purpose Periodic acid-Schiff (PAS) positive patterns of vasculogenic mimicry (VM) have been associated with poor prognosis in uveal melanoma (UM). We examined these patterns with digital image analysis and transmission electron microscopy, and correlated them with BAP-1 expression, gene expression class, macrophage infiltration, and metastatic disease in full tumor cross-sections and intratumor regions. Methods Thirty-two enucleated eyes with UM were stained immunohistochemically (BAP-1, laminin, CD31, and CD68) and with PAS without hematoxylin counterstain. Retrospective data on gene expression class and patient survival were retrieved. Tumor sections were digitally scanned and analyzed with the QuPath Bioimage analysis software, and imaged with transmission electron microscopy. Results The mean area proportion covered by CD31, laminin, and PAS positive patterns in tumor cross-sections was 0.9% (SD 0.6), 3.0% (SD 1.9), and 8.4% (SD 5.9), respectively. PAS density was statistically significantly greater in tumors with gene expression class 2 (p=0.02). The cumulative 5-year metastasis-free survival decreased for each quartile of increased PAS density (1.0, 0.75, 0.40, and 0.17, p=0.004). Forty percent of the tumors had heterogeneous BAP-1 expression. Intratumor regions with low BAP-1 expression were more likely to harbor VM (p<0.0001), and had statistically significantly greater PAS density (p<0.0001) and number of CD68 positive cells (p=0.01). Conclusions PAS positive patterns in UM are composed of a mixture of blood vessels and extracellular matrix (ECM), including VM. Increased density of PAS positive patterns correlated with gene expression class and metastasis, and colocated to tumor regions with macrophage infiltration and low BAP-1 expression.
Collapse
Affiliation(s)
- Gustav Stålhammar
- Ophthalmic Pathology and Oncology Service, St. Erik Eye Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Departments of Ophthalmology and Pathology, Emory University School of Medicine, Atlanta, GA
| | - Thonnie Rose O. See
- Departments of Ophthalmology and Pathology, Emory University School of Medicine, Atlanta, GA
| | - Stephen S. Phillips
- Departments of Ophthalmology and Pathology, Emory University School of Medicine, Atlanta, GA
| | - Hans E. Grossniklaus
- Departments of Ophthalmology and Pathology, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
7
|
Wisely CE, Sayed JA, Tamez H, Zelinka C, Abdel-Rahman MH, Fischer AJ, Cebulla CM. The chick eye in vision research: An excellent model for the study of ocular disease. Prog Retin Eye Res 2017; 61:72-97. [PMID: 28668352 PMCID: PMC5653414 DOI: 10.1016/j.preteyeres.2017.06.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 06/24/2017] [Accepted: 06/27/2017] [Indexed: 02/06/2023]
Abstract
The domestic chicken, Gallus gallus, serves as an excellent model for the study of a wide range of ocular diseases and conditions. The purpose of this manuscript is to outline some anatomic, physiologic, and genetic features of this organism as a robust animal model for vision research, particularly for modeling human retinal disease. Advantages include a sequenced genome, a large eye, relative ease of handling and maintenance, and ready availability. Relevant similarities and differences to humans are highlighted for ocular structures as well as for general physiologic processes. Current research applications for various ocular diseases and conditions, including ocular imaging with spectral domain optical coherence tomography, are discussed. Several genetic and non-genetic ocular disease models are outlined, including for pathologic myopia, keratoconus, glaucoma, retinal detachment, retinal degeneration, ocular albinism, and ocular tumors. Finally, the use of stem cell technology to study the repair of damaged tissues in the chick eye is discussed. Overall, the chick model provides opportunities for high-throughput translational studies to more effectively prevent or treat blinding ocular diseases.
Collapse
Affiliation(s)
- C Ellis Wisely
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, 915 Olentangy River Rd, Columbus, OH 43212, USA
| | - Javed A Sayed
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, 915 Olentangy River Rd, Columbus, OH 43212, USA
| | - Heather Tamez
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, 915 Olentangy River Rd, Columbus, OH 43212, USA
| | - Chris Zelinka
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 West 10th Avenue, Columbus, OH 43210, USA
| | - Mohamed H Abdel-Rahman
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, 915 Olentangy River Rd, Columbus, OH 43212, USA
| | - Andy J Fischer
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 West 10th Avenue, Columbus, OH 43210, USA.
| | - Colleen M Cebulla
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, 915 Olentangy River Rd, Columbus, OH 43212, USA.
| |
Collapse
|
8
|
Abstract
Uveal melanoma (UM), a rare cancer of the eye, is distinct from cutaneous melanoma by its etiology, the mutation frequency and profile, and its clinical behavior including resistance to targeted therapy and immune checkpoint blockers. Primary disease is efficiently controlled by surgery or radiation therapy, but about half of UMs develop distant metastasis mostly to the liver. Survival of patients with metastasis is below 1 year and has not improved in decades. Recent years have brought a deep understanding of UM biology characterized by initiating mutations in the G proteins GNAQ and GNA11. Cytogenetic alterations, in particular monosomy of chromosome 3 and amplification of the long arm of chromosome 8, and mutation of the BRCA1-associated protein 1, BAP1, a tumor suppressor gene, or the splicing factor SF3B1 determine UM metastasis. Cytogenetic and molecular profiling allow for a very precise prognostication that is still not matched by efficacious adjuvant therapies. G protein signaling has been shown to activate the YAP/TAZ pathway independent of HIPPO, and conventional signaling via the mitogen-activated kinase pathway probably also contributes to UM development and progression. Several lines of evidence indicate that inflammation and macrophages play a pro-tumor role in UM and in its hepatic metastases. UM cells benefit from the immune privilege in the eye and may adopt several mechanisms involved in this privilege for tumor escape that act even after leaving the niche. Here, we review the current knowledge of the biology of UM and discuss recent approaches to UM treatment.
Collapse
Affiliation(s)
- Adriana Amaro
- Laboratory of Molecular Pathology, Department of Integrated Oncology Therapies, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, L.go Rosanna Benzi 10, 16132, Genoa, Italy
| | - Rosaria Gangemi
- Laboratory of Biotherapies, Department of Integrated Oncology Therapies, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Francesca Piaggio
- Laboratory of Molecular Pathology, Department of Integrated Oncology Therapies, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, L.go Rosanna Benzi 10, 16132, Genoa, Italy
| | - Giovanna Angelini
- Laboratory of Molecular Pathology, Department of Integrated Oncology Therapies, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, L.go Rosanna Benzi 10, 16132, Genoa, Italy
| | - Gaia Barisione
- Laboratory of Biotherapies, Department of Integrated Oncology Therapies, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Silvano Ferrini
- Laboratory of Biotherapies, Department of Integrated Oncology Therapies, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Ulrich Pfeffer
- Laboratory of Molecular Pathology, Department of Integrated Oncology Therapies, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, L.go Rosanna Benzi 10, 16132, Genoa, Italy.
| |
Collapse
|
9
|
Animal Models of Uveal Melanoma: Methods, Applicability, and Limitations. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4521807. [PMID: 27366747 PMCID: PMC4913058 DOI: 10.1155/2016/4521807] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/08/2016] [Indexed: 02/06/2023]
Abstract
Animal models serve as powerful tools for investigating the pathobiology of cancer, identifying relevant pathways, and developing novel therapeutic agents. They have facilitated rapid scientific progress in many tumor entities. However, for establishing a powerful animal model of uveal melanoma fundamental challenges remain. To date, no animal model offers specific genetic attributes as well as histologic, immunologic, and metastatic features of uveal melanoma. Syngeneic models with intraocular injection of cutaneous melanoma cells may suit best for investigating immunologic/tumor biology aspects. However, differences between cutaneous and uveal melanoma regarding genetics and metastasis remain problematic. Human xenograft models are widely used for evaluating novel therapeutics but require immunosuppression to allow tumor growth. New approaches aim to establish transgenic mouse models of spontaneous uveal melanoma which recently provided preliminary promising results. Each model provides certain benefits and may render them suitable for answering a respective scientific question. However, all existing models also exhibit relevant limitations which may have led to delayed research progress. Despite refined therapeutic options for the primary ocular tumor, patients' prognosis has not improved since the 1970s. Basic research needs to further focus on a refinement of a potent animal model which mimics uveal melanoma specific mechanisms of progression and metastasis. This review will summarise and interpret existing animal models of uveal melanoma including recent advances in the field.
Collapse
|
10
|
Ozaki S, Vuyyuru R, Kageyama K, Terai M, Ohara M, Cheng H, Manser T, Mastrangelo MJ, Aplin AE, Sato T. Establishment and Characterization of Orthotopic Mouse Models for Human Uveal Melanoma Hepatic Colonization. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 186:43-56. [PMID: 26613897 DOI: 10.1016/j.ajpath.2015.09.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 09/07/2015] [Accepted: 09/17/2015] [Indexed: 01/25/2023]
Abstract
Uveal melanoma (UM) is a rare type of melanoma, although it is the most common primary ocular malignant tumor in adults. Nearly one-half the patients with primary UM subsequently develop systemic metastasis, preferentially to the liver. Currently, no treatment is effective for UM hepatic metastasis, and the prognosis is universally poor. The main challenge in designing a treatment strategy for UM hepatic metastasis is the lack of suitable animal models. We developed two orthotopic mouse models for human UM hepatic metastases: direct hepatic implantation model (intrahepatic dissemination model) and splenic-implantation model (hematogenous dissemination model) and investigated the tumorgenesis in the liver. A human UM cell line, established from a hepatic metastasis and nonobese diabetic severe combined immunodeficient γ mice, were used for development of in vivo tumor models. In the direct hepatic implantation model, a localized tumor developed in the liver in all cases and intrahepatic dissemination was subsequently seen in about one-half of cases. However, in the splenic implantation model, multiple hepatic metastases were observed after splenic implantation. Hepatic tumors subsequently seeded intra-abdominal metastasis; however, lung metastases were not seen. These findings are consistent with those observed in human UM hepatic metastases. These orthotopic mouse models offer useful tools to investigate the biological behavior of human UM cells in the liver.
Collapse
Affiliation(s)
- Shinji Ozaki
- Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; Department of Breast Surgery, National Hospital Organization, Kure Medical Center/Chugoku Cancer Center, Kure-shi, Japan
| | - Raja Vuyyuru
- Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ken Kageyama
- Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Mizue Terai
- Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Masahiro Ohara
- Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Hanyin Cheng
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Tim Manser
- Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Michael J Mastrangelo
- Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Andrew E Aplin
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Takami Sato
- Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
11
|
Protein MRI contrast agent with unprecedented metal selectivity and sensitivity for liver cancer imaging. Proc Natl Acad Sci U S A 2015; 112:6607-12. [PMID: 25971726 DOI: 10.1073/pnas.1423021112] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
With available MRI techniques, primary and metastatic liver cancers that are associated with high mortality rates and poor treatment responses are only diagnosed at late stages, due to the lack of highly sensitive contrast agents without Gd(3+) toxicity. We have developed a protein contrast agent (ProCA32) that exhibits high stability for Gd(3+) and a 10(11)-fold greater selectivity for Gd(3+) over Zn(2+) compared with existing contrast agents. ProCA32, modified from parvalbumin, possesses high relaxivities (r1/r2: 66.8 mmol(-1)⋅s(-1)/89.2 mmol(-1)⋅s(-1) per particle). Using T1- and T2-weighted, as well as T2/T1 ratio imaging, we have achieved, for the first time (to our knowledge), robust MRI detection of early liver metastases as small as ∼0.24 mm in diameter, much smaller than the current detection limit of 10-20 mm. Furthermore, ProCA32 exhibits appropriate in vivo preference for liver sinusoidal spaces and pharmacokinetics for high-quality imaging. ProCA32 will be invaluable for noninvasive early detection of primary and metastatic liver cancers as well as for monitoring treatment and guiding therapeutic interventions, including drug delivery.
Collapse
|
12
|
van der Ent W, Burrello C, de Lange MJ, van der Velden PA, Jochemsen AG, Jager MJ, Snaar-Jagalska BE. Embryonic Zebrafish: Different Phenotypes after Injection of Human Uveal Melanoma Cells. Ocul Oncol Pathol 2015; 1:170-81. [PMID: 27171126 DOI: 10.1159/000370159] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 11/26/2014] [Indexed: 12/21/2022] Open
Abstract
Although murine xenograft models for human uveal melanoma (UM) are available, they are of limited utility for screening large compound libraries for the discovery of new drugs. We need new preclinical models which can efficiently evaluate drugs that can treat UM metastases. The zebrafish embryonic model is ideal for drug screening purposes because it allows the investigation of potential antitumor properties of drugs within 1 week. The optical transparency of the zebrafish provides unique possibilities for live imaging of fluorescence-labelled cancer cells and their behavior. In addition, the adaptive immune response, which is responsible for the rejection of transplanted material, is not yet present in the early stages of fish development, and systemic immunosuppression is therefore not required to allow growth of tumor cells. We studied the behavior of UM cells following injection into zebrafish embryos and observed different phenotypes. We also analyzed cell migration, proliferation, formation of micrometastasis and interaction with the host microenvironment. Significant differences were noted between cell lines: cells derived from metastases showed more migration and proliferation than cells derived from the primary tumors. The addition of the c-Met inhibitor crizotinib to the water in which the larvae were kept reduced the migration and proliferation of UM cells expressing c-Met. This indicates the applicability of the zebrafish xenografts for testing novel inhibitory compounds and provides a fast and sensitive in vivo vertebrate model for preclinical drug screening to combat UM.
Collapse
Affiliation(s)
- Wietske van der Ent
- Institute of Biology, Leiden University, Leiden University Medical Center, Leiden, The Netherlands; Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Claudia Burrello
- Institute of Biology, Leiden University, Leiden University Medical Center, Leiden, The Netherlands
| | - Mark J de Lange
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Aart G Jochemsen
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - B Ewa Snaar-Jagalska
- Institute of Biology, Leiden University, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
13
|
Yang H, Cao J, Grossniklaus HE. Uveal Melanoma Metastasis Models. Ocul Oncol Pathol 2015; 1:151-60. [PMID: 27171919 DOI: 10.1159/000370153] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 11/26/2014] [Indexed: 01/12/2023] Open
Abstract
Metastatic disease is the leading cause of death among patients with uveal melanoma. Treatment options for patients with clinically disseminated disease are usually unsuccessful. In vitro and in vivo models are important tools to investigate the pathogenesis of metastatic uveal melanomas and develop treatments for the metastases. In vitro experimental approaches focusing on cell invasion/migration which mimic the steps of the complex metastatic process may also be used for the identification of potential anti-invasion/migration drugs that may inhibit the spreading of tumor cells or the development of metastases. The effects of these drugs must subsequently be confirmed in reliable in vivo models before entering the clinical trial phase. Several models of intraocular melanoma with metastases in rodents and rabbits are currently being used. Most experimental models of uveal melanoma metastases require injection or implantation of melanoma cells into orthotopic locations, including into the liver, spleen, tail vein, or the left ventricle of the heart, in order to mimic the metastatic process.
Collapse
Affiliation(s)
- Hua Yang
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Ga., USA
| | - Jinfeng Cao
- Department of Ophthalmology, LUMC, Leiden, The Netherlands
| | - Hans E Grossniklaus
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Ga., USA
| |
Collapse
|
14
|
Surriga O, Rajasekhar VK, Ambrosini G, Dogan Y, Huang R, Schwartz GK. Crizotinib, a c-Met inhibitor, prevents metastasis in a metastatic uveal melanoma model. Mol Cancer Ther 2013; 12:2817-26. [PMID: 24140933 DOI: 10.1158/1535-7163.mct-13-0499] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Uveal melanoma is the most common primary intraocular malignant tumor in adults and half of the primary tumors will develop fatal metastatic disease to the liver and the lung. Crizotinib, an inhibitor of c-Met, anaplastic lymphoma kinase (ALK), and ROS1, inhibited the phosphorylation of the c-Met receptor but not of ALK or ROS1 in uveal melanoma cells and tumor tissue. Consequently, migration of uveal melanoma cells was suppressed in vitro at a concentration associated with the specific inhibition of c-Met phosphorylation. This effect on cell migration could be recapitulated with siRNA specific to c-Met but not to ALK or ROS1. Therefore, we developed a uveal melanoma metastatic mouse model with EGFP-luciferase-labeled uveal melanoma cells transplanted by retro-orbital injections to test the effect of crizotinib on metastasis. In this model, there was development of melanoma within the eye and also metastases to the liver and lung at 7 weeks after the initial transplantation. When mice were treated with crizotinib starting 1 week after the transplantation, we observed a significant reduction in the development of metastases as compared with untreated control sets. These results indicate that the inhibition of c-Met activity alone may be sufficient to strongly inhibit metastasis of uveal melanoma from forming, suggesting crizotinib as a potential adjuvant therapy for patients with primary uveal melanoma who are at high risk for the development of metastatic disease.
Collapse
Affiliation(s)
- Oliver Surriga
- Corresponding Author: Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065.
| | | | | | | | | | | |
Collapse
|
15
|
Host pigment epithelium-derived factor (PEDF) prevents progression of liver metastasis in a mouse model of uveal melanoma. Clin Exp Metastasis 2013; 30:969-76. [PMID: 23793989 DOI: 10.1007/s10585-013-9596-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 06/04/2013] [Indexed: 12/28/2022]
Abstract
Uveal melanoma (UM) has a 30 % 5-year mortality rate, primarily due to liver metastasis. Both angiogenesis and stromagenesis are important mechanisms for the progression of liver metastasis. Pigment epithelium-derived factor (PEDF), an anti-angiogenic and anti-stromagenic protein, is produced by hepatocytes. Exogenous PEDF suppresses metastasis progression; however, the effects of host-produced PEDF on metastasis progression are unknown. We hypothesize that host PEDF inhibits liver metastasis progression through a mechanism involving angiogenesis and stromagenesis. Mouse melanoma cells were injected into the posterior ocular compartment of PEDF-null mice and control mice. After 1 month, the number, size, and mean vascular density (MVD) of liver metastases were determined. The stromal component of hepatic stellate cells (HSCs) and the type III collagen they produce was evaluated by immunohistochemistry. Host PEDF inhibited the total area of liver metastasis and the frequency of macrometastases (diameter >200 μm) but did not affect the total number of metastases. Mice expressing PEDF exhibited significantly lower MVD and less type III collagen production in metastases. An increase in activated HSCs was seen in the absence of PEDF, but this result was not statistically significant. In conclusion, host PEDF inhibits the progression of hepatic metastases in a mouse model of UM, and loss of PEDF is accompanied by an increase in tumor blood vessel density and type III collagen.
Collapse
|
16
|
Grossniklaus HE. Progression of ocular melanoma metastasis to the liver: the 2012 Zimmerman lecture. JAMA Ophthalmol 2013; 131:462-9. [PMID: 23392528 DOI: 10.1001/jamaophthalmol.2013.2547] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
IMPORTANCE To the best of my knowledge, this study demonstrates for the first time small, apparently dormant micrometastasis in the liver of patients with uveal melanoma. OBJECTIVE To evaluate the histological and immunohistochemical findings in metastatic uveal melanoma to the liver. DESIGN Samples of liver were obtained at autopsy from patients who died of metastatic uveal melanoma to the liver. SETTING L. F. Montgomery Laboratory, Emory Eye Center, Atlanta, Georgia. PARTICIPANTS A total of 10 patients who died of metastatic uveal melanoma to the liver. INTERVENTION Sections of the liver were examined with hematoxylin-eosin, periodic acid-Schiff, Masson trichrome, or reticulin stains. MAIN OUTCOME MEASURES The tumors' morphology, growth pattern, mean vascular density, and mitotic index were determined with the aid of immunohistochemical stains for S100, HMB45, CD31, and Ki67. RESULTS Stage 1 metastases (defined as tumor clusters ≤50 μm in diameter) were identified in the sinusoidal spaces of 9 of 10 patients (90%). Stage 1 metastases were avascular and lacked mitotic activity. Stage 2 metastases (defined as tumors measuring 51-500 μm in diameter) and stage 3 metastases (defined as tumors measuring >500 μm in diameter) were found in all patients. Immunohistochemical stains were positive for S100 or HMB45 in all tumors. Overall, stage 1 metastases outnumbered stage 2 metastases (which outnumbered stage 3 metastases). The mean vascular density and mitotic index increased from stage 2 to stage 3 metastases (P < .05). The architecture of stage 2 metastases mimicked the surrounding hepatic parenchyma, whereas stage 3 metastases exhibited either lobular or portal growth patterns. CONCLUSIONS Uveal melanoma that spreads to the liver can be categorized as stage 1 (≤50 μm in diameter), stage 2 (51-500 μm in diameter), or stage 3 (>500 μm in diameter) metastases. The later stage exhibits a lobular or portal pattern of growth. During this progression, tumors become vascularized and mitotically active.
Collapse
|
17
|
Shimojo Y, Akimoto M, Hisanaga T, Tanaka T, Tajima Y, Honma Y, Takenaga K. Attenuation of reactive oxygen species by antioxidants suppresses hypoxia-induced epithelial-mesenchymal transition and metastasis of pancreatic cancer cells. Clin Exp Metastasis 2012; 30:143-54. [PMID: 22833345 DOI: 10.1007/s10585-012-9519-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 07/09/2012] [Indexed: 12/11/2022]
Abstract
Hypoxia has been shown to promote metastasis of cancer cells through induction of epithelial-mesenchymal transition (EMT). It is also known to cause generation of reactive oxygen species (ROS). We investigated here the role of ROS in hypoxia-induced EMT and whether attenuation of ROS by antioxidants suppresses hypoxia-induced EMT and metastasis of human pancreatic cancer cells in a xenograft nude mouse model. PANC-1 and MiaPaCa-2 cells exposed to hypoxia (1 % O(2)) showed increased ROS generation and characteristic changes of EMT such as morphological changes, enhanced invasiveness, and upregulation of EMT regulators, SLUG, SNAI1 and TWIST. The antioxidants N-acetylcysteine (NAC) and ebselen significantly suppressed EMT and the expression of EMT regulators during hypoxia. NAC abrogated activation of HIF-1α and NF-κB, both of which were found to play an active role in hypoxia-induced EMT. Administration of NAC to nude mice with orthotopic tumors suppressed the expression of EMT regulators in hypoxic areas and significantly inhibited hepatic metastasis. Together, the present findings demonstrate that attenuation of ROS by antioxidants suppresses hypoxia-induced EMT and metastatic phenotype, suggesting that antioxidants may be of therapeutic value in treating pancreatic cancers.
Collapse
Affiliation(s)
- Yoshihide Shimojo
- Department of Digestive and General Surgery, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, Japan
| | | | | | | | | | | | | |
Collapse
|
18
|
Liang Z, Zhan W, Zhu A, Yoon Y, Lin S, Sasaki M, Klapproth JMA, Yang H, Grossniklaus HE, Xu J, Rojas M, Voll RJ, Goodman MM, Arrendale RF, Liu J, Yun CC, Snyder JP, Liotta DC, Shim H. Development of a unique small molecule modulator of CXCR4. PLoS One 2012; 7:e34038. [PMID: 22485156 PMCID: PMC3317778 DOI: 10.1371/journal.pone.0034038] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 02/21/2012] [Indexed: 01/07/2023] Open
Abstract
Background Metastasis, the spread and growth of tumor cells to distant organ sites, represents the most devastating attribute and plays a major role in the morbidity and mortality of cancer. Inflammation is crucial for malignant tumor transformation and survival. Thus, blocking inflammation is expected to serve as an effective cancer treatment. Among anti-inflammation therapies, chemokine modulation is now beginning to emerge from the pipeline. CXC chemokine receptor-4 (CXCR4) and its ligand stromal cell-derived factor-1 (CXCL12) interaction and the resulting cell signaling cascade have emerged as highly relevant targets since they play pleiotropic roles in metastatic progression. The unique function of CXCR4 is to promote the homing of tumor cells to their microenvironment at the distant organ sites. Methodology/Principal Findings We describe the actions of N,N′-(1,4-phenylenebis(methylene))dipyrimidin-2-amine (designated MSX-122), a novel small molecule and partial CXCR4 antagonist with properties quite unlike that of any other reported CXCR4 antagonists, which was prepared in a single chemical step using a reductive amination reaction. Its specificity toward CXCR4 was tested in a binding affinity assay and a ligand competition assay using 18F-labeled MSX-122. The potency of the compound was determined in two functional assays, Matrigel invasion assay and cAMP modulation. The therapeutic potential of MSX-122 was evaluated in three different murine models for inflammation including an experimental colitis, carrageenan induced paw edema, and bleomycin induced lung fibrosis and three different animal models for metastasis including breast cancer micrometastasis in lung, head and neck cancer metastasis in lung, and uveal melanoma micrometastasis in liver in which CXCR4 was reported to play crucial roles. Conclusions/Significance We developed a novel small molecule, MSX-122, that is a partial CXCR4 antagonist without mobilizing stem cells, which can be safer for long-term blockade of metastasis than other reported CXCR4 antagonists.
Collapse
Affiliation(s)
- Zhongxing Liang
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia, United States of America
| | - Weiqiang Zhan
- Department of Chemistry, Emory University, Atlanta, Georgia, United States of America
| | - Aizhi Zhu
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia, United States of America
| | - Younghyoun Yoon
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia, United States of America
| | - Songbai Lin
- Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Maiko Sasaki
- Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | | | - Hua Yang
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States of America
| | - Hans E. Grossniklaus
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States of America
| | - Jianguo Xu
- Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Mauricio Rojas
- Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Ronald J. Voll
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia, United States of America
| | - Mark M. Goodman
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia, United States of America
- Winship Cancer Institute, Emory University, Atlanta, Georgia, United States of America
| | - Richard F. Arrendale
- The Emory Institute for Drug Discovery, Emory University, Atlanta, Georgia, United States of America
| | - Jin Liu
- Department of Chemistry, Emory University, Atlanta, Georgia, United States of America
| | - C. Chris Yun
- Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - James P. Snyder
- Department of Chemistry, Emory University, Atlanta, Georgia, United States of America
- The Emory Institute for Drug Discovery, Emory University, Atlanta, Georgia, United States of America
- * E-mail: (HS); (DCL); (JPS)
| | - Dennis C. Liotta
- Department of Chemistry, Emory University, Atlanta, Georgia, United States of America
- Winship Cancer Institute, Emory University, Atlanta, Georgia, United States of America
- The Emory Institute for Drug Discovery, Emory University, Atlanta, Georgia, United States of America
- * E-mail: (HS); (DCL); (JPS)
| | - Hyunsuk Shim
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia, United States of America
- Winship Cancer Institute, Emory University, Atlanta, Georgia, United States of America
- * E-mail: (HS); (DCL); (JPS)
| |
Collapse
|
19
|
Yang H, Jager MJ, Grossniklaus HE. Bevacizumab suppression of establishment of micrometastases in experimental ocular melanoma. Invest Ophthalmol Vis Sci 2010; 51:2835-42. [PMID: 20089875 PMCID: PMC2874122 DOI: 10.1167/iovs.09-4755] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 12/16/2009] [Accepted: 01/10/2010] [Indexed: 02/06/2023] Open
Abstract
PURPOSE This study was undertaken to determine whether anti-vascular endothelial growth factor (VEGF) therapy inhibits growth of primary uveal melanoma and spread of its hepatic micrometastases. METHODS The human uveal melanoma cell lines Mel290 and Mel 270, HUVECs, mouse B16LS9 melanoma cells, and mouse vascular endothelial cells were separately cultured or co-cultured and incubated with bevacizumab or IgG1. The level of VEGF protein in the culture medium was measured by ELISA. In vitro angiogenesis and invasion assays were performed under bevacizumab or IgG1 treatment. Mel290 or B16LS9 cells were inoculated into NU/NU or C57Bl/6 mouse eyes which were enucleated after 7 days. The sizes of the intraocular tumors were determined. Time and dosage experiments were performed by using 50 or 250 microg bevacizumab starting at day 1 or 4 after inoculation. Hepatic micrometastases were enumerated. Proliferation, apoptosis, and angiogenesis markers were detected in the ocular tumor by immunofluorescence staining. RESULTS Bevacizumab significantly reduced the level of VEGF in the culture media from human uveal melanoma cells, mouse melanoma cells, and co-cultured cells. It also inhibited cell tube formation and decreased in vitro invasion of tumor cells. In the mouse model, bevacizumab suppressed primary ocular melanoma growth and the formation of hepatic micrometastases in a dose-dependent manner. Furthermore, immunohistochemical staining showed decreased Ki67 and unchanged caspase 3 expression after treatment with bevacizumab. CONCLUSIONS Treatment with bevacizumab suppressed in vitro growth and in vivo hepatic micrometastasis of ocular melanoma cells. Bevacizumab is a potential therapeutic agent for the treatment of uveal melanoma micrometastases.
Collapse
MESH Headings
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Apoptosis
- Bevacizumab
- Caspase 3/metabolism
- Cell Proliferation
- Coculture Techniques
- Dose-Response Relationship, Drug
- Enzyme-Linked Immunosorbent Assay
- Female
- Humans
- Ki-67 Antigen/metabolism
- Liver Neoplasms/blood supply
- Liver Neoplasms/drug therapy
- Liver Neoplasms/secondary
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/secondary
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/metabolism
- Tumor Cells, Cultured
- Uveal Neoplasms/blood supply
- Uveal Neoplasms/drug therapy
- Uveal Neoplasms/pathology
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Hua Yang
- From the Departments of Ophthalmology and
| | - Martine J. Jager
- the Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans E. Grossniklaus
- From the Departments of Ophthalmology and
- Pathology, Emory University School of Medicine, Atlanta, Georgia; and
| |
Collapse
|
20
|
Némati F, Sastre-Garau X, Laurent C, Couturier J, Mariani P, Desjardins L, Piperno-Neumann S, Lantz O, Asselain B, Plancher C, Robert D, Péguillet I, Donnadieu MH, Dahmani A, Bessard MA, Gentien D, Reyes C, Saule S, Barillot E, Roman-Roman S, Decaudin D. Establishment and characterization of a panel of human uveal melanoma xenografts derived from primary and/or metastatic tumors. Clin Cancer Res 2010; 16:2352-62. [PMID: 20371695 DOI: 10.1158/1078-0432.ccr-09-3066] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Uveal melanoma is the most common primary intraocular malignant tumor in adults and is defined by a poor natural outcome, as 50% of patients die from metastases. The aim of this study was to develop and characterize a panel of human uveal melanoma xenografts transplanted into immunodeficient mice. EXPERIMENTAL DESIGN Ninety tumor specimens were grafted into severe combined immunodeficient mice, and 25 transplantable xenografts were then established (28%). Relationship between tumor graft and clinical, biological, and therapeutic features of the patients included were investigated. Characterization of 16 xenografts included histology, molecular analyses by immunohistochemistry, genetic alteration analysis (single-nucleotide polymorphism), and specific tumor antigen expression by quantitative reverse transcription-PCR. Pharmacologic characterization (chemosensitivity) was also done in four models using two drugs, temozolomide and fotemustine, currently used in the clinical management of uveal melanoma. RESULTS Take rate of human uveal melanoma was 28% (25 of 90). Tumor take was independent of size, histologic parameters, or chromosome 3 monosomy but was significantly higher in metastatic tumors. Interestingly, in vivo tumor growth was prognostic for a lower metastasis-free survival in patients with primary tumors. A high concordance between the patients' tumors and their corresponding xenografts was found for all parameters tested (histology, genetic profile, and tumor antigen expression). Finally, the four xenografts studied displayed different response profiles to chemotherapeutic agents. CONCLUSIONS Based on these results, this panel of 16 uveal melanoma xenografts represents a useful preclinical tool for both pharmacologic and biological assessments.
Collapse
Affiliation(s)
- Fariba Némati
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
A mutated EGFR is sufficient to induce malignant melanoma with genetic background-dependent histopathologies. J Invest Dermatol 2010; 130:249-58. [PMID: 19609310 DOI: 10.1038/jid.2009.213] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Melanoma is a tumor with a very low cure rate once metastasized. Although many genes important for melanoma induction, transformation, and metastasis have been identified, the process of melanomagenesis is only partly understood. Melanoma mediators are easiest to investigate in cell culture models, but animal models are required to evaluate their importance in the context of the whole organism. Here, we describe a transgenic melanoma model in medaka. The oncogenic receptor tyrosine kinase, Xmrk, responsible for melanoma formation in Xiphophorus, was stably expressed under the control of a pigment cell-specific promoter. The transgenic fish developed pigment cell tumors with a penetrance of 100%. The model was used for monitoring the in vivo relevance of several apoptosis and differentiation genes, and for induction of melanoma-relevant signal transduction pathways. We found that Stat5 activation, and Mitf and Bcl-2 levels correlated with a more aggressive stage of the malignancy. Interestingly, different types of pigment cell tumors occurred depending on the genetic background, namely invasive melanoma, uveal melanoma, or exophytic and less aggressive pigment cell tumors called xanthoerythrophoroma. Furthermore, on p53 mutant background, the expression of xmrk led to the appearance of giant focal pigment cell tumors, whereas tumor onset was unchanged compared with wild-type medaka.
Collapse
|
22
|
Yang H, Grossniklaus HE. Constitutive overexpression of pigment epithelium-derived factor inhibition of ocular melanoma growth and metastasis. Invest Ophthalmol Vis Sci 2010; 51:28-34. [PMID: 19661223 PMCID: PMC2819011 DOI: 10.1167/iovs.09-4138] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 07/14/2009] [Accepted: 07/30/2009] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Pigment epithelium-derived factor (PEDF) is known to be an angiogenesis suppressor and to have antitumor effects. This study investigates whether constitutive overexpression of PEDF inhibits the growth and hepatic micrometastasis of ocular melanoma. METHODS Real-time RT-PCR was used to detect endogenous PEDF expression in human uveal melanoma cell lines and mouse melanoma cells. A lentiviral vector containing a mouse PEDF expression sequence was constructed and transduced into mouse melanoma cells in vitro. Transgene expression was assessed by Western blot analysis. Angiogenesis and transendothelial migration assays were performed in constitutively stable PEDF-overexpressing cells and transduced lentiviral vector control cells. The size and microvessel density of the ocular tumor and the number of hepatic micrometastasis were compared between the mice inoculated with PEDF-overexpressing tumor cells and those mice with the control cell line. RESULTS Four human uveal melanoma and three mouse melanoma cell lines were found to express PEDF mRNA. Endogenous overexpressing PEDF melanoma cells lost the ability to migrate and form tubes in vitro. In the animal experiment, the size of the ocular melanoma and the number of hepatic micrometastasis were decreased and microvessel density was also reduced in mice inoculated with constitutively overexpressing PEDF melanoma cells. CONCLUSIONS Lentivirus-mediated gene transfer of PEDF decreased the growth of ocular melanoma and its hepatic micrometastasis in a mouse ocular melanoma model. Dual antitumor/antiangiogenic activities of PEDF suggest that PEDF gene therapy may be considered an approach for the treatment of ocular melanoma.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Eye Proteins/genetics
- Fluorescent Antibody Technique, Indirect
- Gene Expression Regulation, Neoplastic/physiology
- Genetic Therapy
- Genetic Vectors
- Humans
- Lentivirus/genetics
- Liver Neoplasms/genetics
- Liver Neoplasms/prevention & control
- Liver Neoplasms/secondary
- Melanoma/blood supply
- Melanoma/genetics
- Melanoma/prevention & control
- Melanoma/secondary
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/genetics
- Melanoma, Experimental/prevention & control
- Melanoma, Experimental/secondary
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neovascularization, Pathologic/prevention & control
- Nerve Growth Factors/genetics
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Serpins/genetics
- Transduction, Genetic
- Transfection
- Tumor Cells, Cultured
- Uveal Neoplasms/blood supply
- Uveal Neoplasms/genetics
- Uveal Neoplasms/pathology
- Uveal Neoplasms/prevention & control
Collapse
Affiliation(s)
- Hua Yang
- From the Departments of Ophthalmology and
| | - Hans E. Grossniklaus
- From the Departments of Ophthalmology and
- Pathology and
- the Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
23
|
Folberg R, Kadkol SS, Frenkel S, Valyi-Nagy K, Jager MJ, Pe'er J, Maniotis AJ. Authenticating cell lines in ophthalmic research laboratories. Invest Ophthalmol Vis Sci 2008; 49:4697-701. [PMID: 18689700 PMCID: PMC2576485 DOI: 10.1167/iovs.08-2324] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Affiliation(s)
- Robert Folberg
- Oakland University William Beaumont School of Medicine, Rochester, Michigan 48309-4401, USA.
| | | | | | | | | | | | | |
Collapse
|