1
|
Yenisehirli G, Borges S, Braun SS, Zuniga AN, Quintana GI, Kutsnetsoff JN, Rodriguez S, Adis EV, Lopez S, Dollar JJ, Stathias V, Volmar CH, Karaca E, Brothers S, Bilbao D, Harbour JW, Correa ZM, Kurtenbach S. Identification of targetable epigenetic vulnerabilities in uveal melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617464. [PMID: 39416076 PMCID: PMC11482939 DOI: 10.1101/2024.10.11.617464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Uveal melanoma (UM) is the most prevalent primary intraocular malignancy in adults, which preferentially metastasizes to the liver in approximately half of all cases. Metastatic UM is notoriously resistant to therapy and is almost uniformly fatal. UM metastasis is most strongly associated with mutational inactivation of the BAP1 tumor suppressor gene. Given the role of BAP1 in epigenetic regulation as the ubiquitin hydrolase subunit of the polycomb repressive deubiquitinase (PR-DUB) complex, we conducted high-throughput drug screening using a well-characterized epigenetic compound library to identify new therapeutic vulnerabilities. We identified several promising new lead compounds, in particular the extra-terminal domain protein (BET) inhibitor mivebresib (ABBV-075). Mivebresib significantly improved survival rates in a metastatic uveal melanoma xenograft mouse model and entirely prevented detectable metastases to the bones, spinal cord, and brain. RNA sequencing revealed a notable overlap between the genes and pathways affected by HDAC and BET inhibition, including the reversal of gene signatures linked to high metastatic risk and upregulation of genes associated with a neuronal phenotype. Together, we found that UM cells are particularly vulnerable to class I HDAC and BET inhibition, and highlight the BET inhibitor mivebresib as a promising candidate for further clinical evaluation.
Collapse
Affiliation(s)
- Gulum Yenisehirli
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Sebastian Borges
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Steffanie S. Braun
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Ashley N. Zuniga
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Gabriela I. Quintana
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Jeffim N. Kutsnetsoff
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Sara Rodriguez
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Emily V. Adis
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Sofia Lopez
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - James J. Dollar
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Vasileios Stathias
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
| | - Claude H. Volmar
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine
| | - Efe Karaca
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine
| | - Shaun Brothers
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine
| | - Daniel Bilbao
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine
| | - J. William Harbour
- Department of Ophthalmology and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center
| | - Zelia M. Correa
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Stefan Kurtenbach
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| |
Collapse
|
2
|
Decaudin D, Némati F, Masliah Planchon J, Seguin-Givelet A, Lefevre M, Etienne V, Ahnine H, Peretti Q, Sourd L, El-Botty R, Huguet L, Lagha S, Hegarat N, Roman-Roman S, Bièche I, Girard N, Montaudon E. Evaluation of Combined Chemotherapy and Genomic-Driven Targeted Therapy in Patient-Derived Xenografts Identifies New Therapeutic Approaches in Squamous Non-Small-Cell Lung Cancer Patients. Cancers (Basel) 2024; 16:2785. [PMID: 39199558 PMCID: PMC11352497 DOI: 10.3390/cancers16162785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
The combination of chemotherapy and targeted therapy has been validated in non-small-cell lung cancer (NSCLC) patients with EGFR mutations. We therefore investigated whether this type of combined approach could be more widely used by targeting other genetic alterations present in NSCLC. PDXs were generated from patients with NSCLC adenocarcinomas (ADCs) and squamous-cell carcinomas (SCCs). Targeted NGS analyses identified various molecular abnormalities in the MAPK and PI3K pathways and in the cell cycle process in our PDX panel. The antitumor efficacy of targeted therapies alone or in combination with chemotherapy was then tested in vivo. We observed that trametinib, BKM120, AZD2014 and palbociclib increased the efficacy of each chemotherapy in SCC PDXs, in contrast to a non-insignificant or slight improvement in ADCs. Furthermore, we observed high efficacy of trametinib in KRAS-, HRAS- and NRAS-mutated tumors (ADCs and SCCs), suggesting that the MEK inhibitor may be useful in a wider population of NSCLC patients, not just those with KRAS-mutated ADCs. Our results suggest that the detection of pathogenic variants by NGS should be performed in all NSCLCs, and particularly in SCCs, to offer patients a more effective combination of chemotherapy and targeted therapy.
Collapse
Affiliation(s)
- Didier Decaudin
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
- Department of Medical Oncology, Institut Curie, 75005 Paris, France; (S.L.); (N.H.); (N.G.)
| | - Fariba Némati
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| | | | - Agathe Seguin-Givelet
- Department of Thoracic Surgery, Curie-Montsouris Thorax Institute, Institut Mutualiste Montsouris, 75014 Paris, France;
- Faculty of Medicine SMBH, Paris 13 University, Sorbonne Paris Cité, 75013 Bobigny, France
| | - Marine Lefevre
- Department of Pathology, Institut Mutualiste Montsouris, 75014 Paris, France;
| | - Vesnie Etienne
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| | - Harry Ahnine
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| | - Quentin Peretti
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| | - Laura Sourd
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| | - Rania El-Botty
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| | - Lea Huguet
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| | - Sarah Lagha
- Department of Medical Oncology, Institut Curie, 75005 Paris, France; (S.L.); (N.H.); (N.G.)
| | - Nadia Hegarat
- Department of Medical Oncology, Institut Curie, 75005 Paris, France; (S.L.); (N.H.); (N.G.)
| | - Sergio Roman-Roman
- Department of Translationnal Research, Institut Curie, PSL University Paris, 75006 Paris, France;
| | - Ivan Bièche
- Department of Genetic, Institut Curie, 75005 Paris, France; (J.M.P.); (I.B.)
| | - Nicolas Girard
- Department of Medical Oncology, Institut Curie, 75005 Paris, France; (S.L.); (N.H.); (N.G.)
- Paris Saclay University, University of Versailles Saint-Quentin-en-Yvelines (UVSQ), 91405 Versailles, France
| | - Elodie Montaudon
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| |
Collapse
|
3
|
Cocco E, de Stanchina E. Patient-Derived-Xenografts in Mice: A Preclinical Platform for Cancer Research. Cold Spring Harb Perspect Med 2024; 14:a041381. [PMID: 37696659 PMCID: PMC11216185 DOI: 10.1101/cshperspect.a041381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
The use of patient-derived xenografts (PDXs) has dramatically improved drug development programs. PDXs (1) reproduce the pathological features and the genomic profile of the parental tumors more precisely than other preclinical models, and (2) more faithfully predict therapy response. However, PDXs have limitations. These include the inability to completely capture tumor heterogeneity and the role of the immune system, the low engraftment efficiency of certain tumor types, and the consequences of the human-host interactions. Recently, the use of novel mouse strains and specialized engraftment techniques has enabled the generation of "humanized" PDXs, partially overcoming such limitations. Importantly, establishing, characterizing, and maintaining PDXs is costly and requires a significant regulatory, administrative, clinical, and laboratory infrastructure. In this review, we will retrace the historical milestones that led to the implementation of PDXs for cancer research, review the most recent innovations in the field, and discuss future avenues to tackle deficiencies that still exist.
Collapse
Affiliation(s)
- Emiliano Cocco
- University of Miami, Miller School of Medicine, Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miami, Florida 33136, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
4
|
Goyeneche AA, Lasiste JME, Abdouh M, Bustamante P, Burnier JV, Burnier MN. Delineating three-dimensional behavior of uveal melanoma cells under anchorage independent or dependent conditions. Cancer Cell Int 2024; 24:180. [PMID: 38783299 PMCID: PMC11118898 DOI: 10.1186/s12935-024-03350-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Although rare, uveal melanoma (UM) is a life-threatening malignancy. Understanding its biology is necessary to improve disease outcome. Three-dimensional (3D) in vitro culture methods have emerged as tools that incorporate physical and spatial cues that better mimic tumor biology and in turn deliver more predictive preclinical data. Herein, we comprehensively characterize UM cells under different 3D culture settings as a suitable model to study tumor cell behavior and therapeutic intervention. METHODS Six UM cell lines were tested in two-dimensional (2D) and 3D-culture conditions. For 3D cultures, we used anchorage-dependent (AD) methods where cells were embedded or seeded on top of basement membrane extracts and anchorage-free (AF) methods where cells were seeded on agarose pre-coated plates, ultra-low attachment plates, and on hanging drops, with or without methylcellulose. Cultures were analyzed for multicellular tumor structures (MCTs) development by phase contrast and confocal imaging, and cell wellbeing was assessed based on viability, membrane integrity, vitality, apoptotic features, and DNA synthesis. Vascular endothelial growth factor (VEGF) production was evaluated under hypoxic conditions for cell function analysis. RESULTS UM cells cultured following anchorage-free methods developed MCTs shaped as spheres. Regardless of their sizes and degree of compaction, these spheres displayed an outer ring of viable and proliferating cells, and a core with less proliferating and apoptotic cells. In contrast, UM cells maintained under anchorage-dependent conditions established several morphological adaptations. Some remained isolated and rounded, formed multi-size irregular aggregates, or adopted a 2D-like flat appearance. These cells invariably conserved their metabolic activity and conserved melanocytic markers (i.e., expression of Melan A/Mart-1 and HMB45). Notably, under hypoxia, cells maintained under 3D conditions secrete more VEGF compared to cells cultured under 2D conditions. CONCLUSIONS Under an anchorage-free environment, UM cells form sphere-like MCTs that acquire attributes reminiscent of abnormal vascularized solid tumors. UM cells behavior in anchorage-dependent manner exposed diverse cells populations in response to cues from an enriched extracellular matrix proteins (ECM) environment, highlighting the plasticity of UM cells. This study provides a 3D cell culture platform that is more predictive of the biology of UM. The integration of such platforms to explore mechanisms of ECM-mediated tumor resistance, metastatic abilities, and to test novel therapeutics (i.e., anti-angiogenics and immunomodulators) would benefit UM care.
Collapse
Affiliation(s)
- Alicia A Goyeneche
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, Research Institute of the McGill University Health Centre, Montreal, Canada.
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Canada.
- Experimental Pathology Unit, Department of Pathology, McGill University, Montreal, Canada.
| | - Jade M E Lasiste
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Mohamed Abdouh
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, Research Institute of the McGill University Health Centre, Montreal, Canada
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Prisca Bustamante
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Julia V Burnier
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, Research Institute of the McGill University Health Centre, Montreal, Canada
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Experimental Pathology Unit, Department of Pathology, McGill University, Montreal, Canada
- Department of Oncology, McGill University, Montreal, Canada
| | - Miguel N Burnier
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, Research Institute of the McGill University Health Centre, Montreal, Canada
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Experimental Pathology Unit, Department of Pathology, McGill University, Montreal, Canada
- Department of Oncology, McGill University, Montreal, Canada
| |
Collapse
|
5
|
Herwig-Carl MC, Sharma A, Tischler V, Pelusi N, Loeffler KU, Holz FG, Zeschnigk M, Landreville S, Auw-Haedrich C, Noberini R, Bonaldi T. Mass Spectrometry-Based Profiling of Histone Post-Translational Modifications in Uveal Melanoma Tissues, Human Melanocytes, and Uveal Melanoma Cell Lines - A Pilot Study. Invest Ophthalmol Vis Sci 2024; 65:27. [PMID: 38349785 PMCID: PMC10868634 DOI: 10.1167/iovs.65.2.27] [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: 07/17/2023] [Accepted: 01/31/2024] [Indexed: 02/15/2024] Open
Abstract
Purpose Epigenetic alterations in uveal melanoma (UM) are still neither well characterized, nor understood. In this pilot study, we sought to provide a deeper insight into the possible role of epigenetic alterations in the pathogenesis of UM and their potential prognostic relevance. To this aim, we comprehensively profiled histone post-translational modifications (PTMs), which represent epigenetic features regulating chromatin accessibility and gene transcription, in UM formalin-fixed paraffin-embedded (FFPE) tissues, control tissues, UM cell lines, and healthy melanocytes. Methods FFPE tissues of UM (n = 24), normal choroid (n = 4), human UM cell lines (n = 7), skin melanocytes (n = 6), and uveal melanocytes (n = 2) were analyzed through a quantitative liquid chromatography-mass spectrometry (LC-MS) approach. Results Hierarchical clustering showed a clear separation with several histone PTMs that changed significantly in a tumor compared to normal samples, in both tissues and cell lines. In addition, several acetylations and H4K20me1 showed lower levels in BAP1 mutant tumors. Some of these changes were also observed when we compared GNA11 mutant tumors with GNAQ tumors. The epigenetic profiling of cell lines revealed that the UM cell lines MP65 and UPMM1 have a histone PTM pattern closer to the primary tissues than the other cell lines analyzed. Conclusions Our results suggest the existence of different histone PTM patterns that may be important for diagnosis and prognosis in UM. However, further analyses are needed to confirm these findings in a larger cohort. The epigenetic characterization of a panel of UM cell lines suggested which cellular models are more suitable for epigenetic investigations.
Collapse
Affiliation(s)
- Martina C. Herwig-Carl
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
- Division of Ophthalmic Pathology, University Hospital Bonn, Bonn, Germany
- Centrum for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
| | - Amit Sharma
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Verena Tischler
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Natalie Pelusi
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Karin U. Loeffler
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
- Division of Ophthalmic Pathology, University Hospital Bonn, Bonn, Germany
| | - Frank G. Holz
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
- Centrum for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
| | - Michael Zeschnigk
- Institute of Human Genetics, University Hospital Essen, Essen, Germany
| | - Solange Landreville
- Department of Ophthalmology and Otolaryngology-Cervicofacial Surgery, Université Laval, Quebec City, Quebec, Canada
| | | | - Roberta Noberini
- Department of Experimental Oncology, European Institute of Oncology (IEO) IRCCS, Milan, Italy
| | - Tiziana Bonaldi
- Department of Experimental Oncology, European Institute of Oncology (IEO) IRCCS, Milan, Italy
- Department of Oncology and Haematology-Oncology (DIPO), University of Milan, Milan, Italy
| |
Collapse
|
6
|
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
|
7
|
Nemati F, de Koning L, Gentien D, Assayag F, Henry E, Ait Rais K, Pierron G, Mariani O, Nijnikoff M, Champenois G, Nicolas A, Meseure D, Gardrat S, Servant N, Hupé P, Kamal M, Le Tourneau C, Piperno-Neumann S, Rodrigues M, Roman-Roman S, Decaudin D, Mariani P, Cassoux N. Patient Derived Xenografts (PDX) Models as an Avatar to Assess Personalized Therapy Options in Uveal Melanoma: A Feasibility Study. Curr Oncol 2023; 30:9090-9103. [PMID: 37887557 PMCID: PMC10604955 DOI: 10.3390/curroncol30100657] [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: 08/14/2023] [Revised: 09/13/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
Uveal melanoma is the most common primary intraocular malignancy in adults. Up to 50% of UM patients develop metastatic disease, usually in the liver. When metastatic, the prognosis is poor, and few treatment options exist. Here, we investigated the feasibility of establishing patient-derived xenografts (PDXs) from a patient's tumor in order to screen for therapies that the patient could benefit from. Samples obtained from 29 primary tumors and liver metastases of uveal melanoma were grafted into SCID mice. PDX models were successfully established for 35% of primary patient tumors and 67% of liver metastases. The tumor take rate was proportional to the risk of metastases. PDXs showed the same morphology, the same GNAQ/11, BAP1, and SF3B1 mutations, and the same chromosome 3 and 8q status as the corresponding patient samples. Six PDX models were challenged with two compounds for 4 weeks. We show that, for 31% of patients with high or intermediate risk of metastasis, the timing to obtain efficacy results on PDX models derived from their primary tumors was compatible with the selection of the therapy to treat the patient after relapse. PDXs could thus be a valid tool ("avatar") to select the best personalized therapy for one third of patients that are most at risk of relapse.
Collapse
Affiliation(s)
- Fariba Nemati
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University Paris, 26 rue d’Ulm, CEDEX 05, 75248 Paris, France
| | - Leanne de Koning
- Translational Research Department, Institut Curie, PSL University Paris, 75248 Paris, France; (L.d.K.)
| | - David Gentien
- Genomics Platform, Translational Research Department, Institut Curie, PSL Research University, 75248 Paris, France
| | - Franck Assayag
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University Paris, 26 rue d’Ulm, CEDEX 05, 75248 Paris, France
| | - Emilie Henry
- Genomics Platform, Translational Research Department, Institut Curie, PSL Research University, 75248 Paris, France
| | - Khadija Ait Rais
- Department of Genetics, Institut Curie, PSL Research University, 75248 Paris, France
| | - Gaelle Pierron
- Department of Genetics, Institut Curie, PSL Research University, 75248 Paris, France
| | - Odette Mariani
- Biological Resource Center, Department of Pathology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Michèle Nijnikoff
- Biological Resource Center, Department of Pathology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Gabriel Champenois
- Department of Biopathology, Institut Curie, PSL Research University, 75248 Paris, France
| | - André Nicolas
- Department of Biopathology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Didier Meseure
- Department of Biopathology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Sophie Gardrat
- Department of Biopathology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Nicolas Servant
- Institut Curie, INSERM U900, CBIO-Centre for Computational Biology, Mines Paris Tech, PSL-Research University, 75248 Paris, France
| | - Philippe Hupé
- Institut Curie, INSERM U900, CBIO-Centre for Computational Biology, Mines Paris Tech, PSL-Research University, 75248 Paris, France
| | - Maud Kamal
- Department of Drug Development and Innovation (D3i), Institut Curie, 75248 Paris, France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, 75248 Paris, France
- INSERM U900 Research Unit, Institut Curie, 92064 Saint-Cloud, France
- Paris-Saclay University, 75248 Paris, France
| | - Sophie Piperno-Neumann
- Department of Medical Oncology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Manuel Rodrigues
- Department of Medical Oncology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Sergio Roman-Roman
- Translational Research Department, Institut Curie, PSL University Paris, 75248 Paris, France; (L.d.K.)
| | - Didier Decaudin
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University Paris, 26 rue d’Ulm, CEDEX 05, 75248 Paris, France
- Department of Medical Oncology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Pascale Mariani
- Department of Surgical Oncology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Nathalie Cassoux
- Department of Oncological Ophthalmology, Institut Curie, Université Paris Cité, 75248 Paris, France
| |
Collapse
|
8
|
Gentien D, Saberi-Ansari E, Servant N, Jolly A, de la Grange P, Némati F, Liot G, Saule S, Teissandier A, Bourc'his D, Girard E, Wong J, Masliah-Planchon J, Narmanli E, Liu Y, Torun E, Goulancourt R, Rodrigues M, Gaudé LV, Reyes C, Bazire M, Chenegros T, Henry E, Rapinat A, Bohec M, Baulande S, M'kacher R, Jeandidier E, Nicolas A, Ciriello G, Margueron R, Decaudin D, Cassoux N, Piperno-Neumann S, Stern MH, Gibcus JH, Dekker J, Heard E, Roman-Roman S, Waterfall JJ. Multi-omics comparison of malignant and normal uveal melanocytes reveals molecular features of uveal melanoma. Cell Rep 2023; 42:113132. [PMID: 37708024 PMCID: PMC10598242 DOI: 10.1016/j.celrep.2023.113132] [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: 01/12/2023] [Revised: 07/10/2023] [Accepted: 08/30/2023] [Indexed: 09/16/2023] Open
Abstract
Uveal melanoma (UM) is a rare cancer resulting from the transformation of melanocytes in the uveal tract. Integrative analysis has identified four molecular and clinical subsets of UM. To improve our molecular understanding of UM, we performed extensive multi-omics characterization comparing two aggressive UM patient-derived xenograft models with normal choroidal melanocytes, including DNA optical mapping, specific histone modifications, and DNA topology analysis using Hi-C. Our gene expression and cytogenetic analyses suggest that genomic instability is a hallmark of UM. We also identified a recurrent deletion in the BAP1 promoter resulting in loss of expression and associated with high risk of metastases in UM patients. Hi-C revealed chromatin topology changes associated with the upregulation of PRAME, an independent prognostic biomarker in UM, and a potential therapeutic target. Our findings illustrate how multi-omics approaches can improve our understanding of tumorigenesis and reveal two distinct mechanisms of gene expression dysregulation in UM.
Collapse
Affiliation(s)
- David Gentien
- Translational Research Department, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France; Genomics Platform, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France.
| | - Elnaz Saberi-Ansari
- Translational Research Department, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France; INSERM U830, Research Center, Institut Curie, PSL Research University, 75005 Paris, France
| | | | | | | | - Fariba Némati
- Translational Research Department, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France; Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, 75248 Paris, France
| | - Géraldine Liot
- Institut Curie, PSL Research University, CNRS, INSERM, UMR3347, U1021, Orsay, France
| | - Simon Saule
- Institut Curie, PSL Research University, CNRS, INSERM, UMR3347, U1021, Orsay, France; Université Paris-Saclay Centre National de La Recherche Scientifique, UMR 3347, Unité 1021, Orsay, France
| | - Aurélie Teissandier
- Institut Curie, PSL Research University, Sorbonne University, INSERM U934, CNRS UMR 3215, 75005 Paris, France
| | - Deborah Bourc'his
- Institut Curie, PSL Research University, Sorbonne University, INSERM U934, CNRS UMR 3215, 75005 Paris, France
| | | | - Jennifer Wong
- Department of Diagnostic and Theranostic Molecular Pathology, Unit of Somatic Genetic, Hospital, Institut Curie, Paris, France
| | - Julien Masliah-Planchon
- Department of Diagnostic and Theranostic Molecular Pathology, Unit of Somatic Genetic, Hospital, Institut Curie, Paris, France
| | - Erkan Narmanli
- Translational Research Department, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France; INSERM U830, Research Center, Institut Curie, PSL Research University, 75005 Paris, France
| | - Yuanlong Liu
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland; Swiss Cancer Center Leman, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Emma Torun
- Institut Curie, PSL Research University, Sorbonne University, INSERM U934, CNRS UMR 3215, 75005 Paris, France
| | | | - Manuel Rodrigues
- Department of Medical Oncology, Institut Curie, PSL Research University, 75005 Paris, France; INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe Labellisée par la Ligue Nationale Contre le Cancer, Department of Genetics, Institut Curie, PSL Research University, 75005 Paris, France
| | - Laure Villoing Gaudé
- Translational Research Department, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France; Genomics Platform, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France
| | - Cécile Reyes
- Translational Research Department, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France; Genomics Platform, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France
| | - Matéo Bazire
- Translational Research Department, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France; Genomics Platform, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France
| | - Thomas Chenegros
- Translational Research Department, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France; Genomics Platform, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France
| | - Emilie Henry
- Translational Research Department, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France; Genomics Platform, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France
| | - Audrey Rapinat
- Translational Research Department, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France; Genomics Platform, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France
| | - Mylene Bohec
- Institut Curie Genomics of Excellence (ICGex) Platform, Institut Curie Research Center, PSL Research University, Paris, France
| | - Sylvain Baulande
- Institut Curie Genomics of Excellence (ICGex) Platform, Institut Curie Research Center, PSL Research University, Paris, France
| | | | - Eric Jeandidier
- Laboratoire de Génétique, Groupe Hospitalier de la Région de Mulhouse Sud-Alsace, Mulhouse, France
| | - André Nicolas
- Pathex, Institut Curie, PSL Research University, Paris, France
| | - Giovanni Ciriello
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland; Swiss Cancer Center Leman, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Raphael Margueron
- Institut Curie, PSL Research University, Sorbonne University, INSERM U934, CNRS UMR 3215, 75005 Paris, France
| | - Didier Decaudin
- Translational Research Department, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France; Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, 75248 Paris, France
| | - Nathalie Cassoux
- Department of Medical Oncology, Institut Curie, PSL Research University, 75005 Paris, France; Department of Ocular Oncology, Faculty of Medicine, Institut Curie, Université de Paris Descartes, 75005 Paris, France
| | - Sophie Piperno-Neumann
- Department of Medical Oncology, Institut Curie, PSL Research University, 75005 Paris, France
| | - Marc-Henri Stern
- INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe Labellisée par la Ligue Nationale Contre le Cancer, Department of Genetics, Institut Curie, PSL Research University, 75005 Paris, France
| | - Johan Harmen Gibcus
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Job Dekker
- Howard Hughes Medical Institute, Department of Systems Biology, Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Edith Heard
- Director's Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Sergio Roman-Roman
- Translational Research Department, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France.
| | - Joshua J Waterfall
- Translational Research Department, Research Center, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 75005 Paris, France; INSERM U830, Research Center, Institut Curie, PSL Research University, 75005 Paris, France.
| |
Collapse
|
9
|
Jin B, Yang L, Ye Q, Pan J. Ferroptosis induced by DCPS depletion diminishes hepatic metastasis in uveal melanoma. Biochem Pharmacol 2023; 213:115625. [PMID: 37245534 DOI: 10.1016/j.bcp.2023.115625] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Hepatic metastasis develops in ∼50% of uveal melanoma (UM) patients with scarcely effective treatment resulting in lethality. The underlying mechanism of liver metastasis remains elusive. Ferroptosis, a cell death form characterized by lipid peroxide, in cancer cells may decrease metastatic colonization. In the present study, we hypothesized that decapping scavenger enzymes (DCPS) impact ferroptosis by regulating mRNA decay during the metastatic colonization of UM cells to liver. We found that inhibition of DCPS by shRNA or RG3039 induced gene transcript alteration and ferroptosis through reducing the mRNA turnover of GLRX. Ferroptosis induced by DCPS inhibition eliminates cancer stem-like cells in UM. Inhibition of DCPS hampered the growth and proliferation both in vitro and in vivo. Furthermore, targeting DCPS diminished hepatic metastasis of UM cells. These findings may shed light on the understanding of DCPS-mediated pre-mRNA metabolic pathway in UM by which disseminated cells gain enhanced malignant features to promote hepatic metastasis, providing a rational target for metastatic colonization in UM.
Collapse
Affiliation(s)
- Bei Jin
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Luo Yang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Qianyun Ye
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
10
|
Aschero R, Castillo-Ecija H, Baulenas-Farres M, Resa-Pares C, Jimenez-Cabaco A, Rodriguez E, Monterrubio C, Perez-Jaume S, Suñol M, Chantada GL, Lavarino C, Mora J, Carcaboso AM. Prognostic value of xenograft engraftment in patients with metastatic high-risk neuroblastoma. Pediatr Blood Cancer 2023; 70:e30318. [PMID: 36973999 DOI: 10.1002/pbc.30318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND Successful engraftment of human cancer biopsies in immunodeficient mice correlates with the poor prognosis of patients. This was reported 30 years ago for children with neuroblastoma, but the standard of care treatment evolved significantly during the last 15 years, leading to improved survival of these patients. Here, we evaluated the association of patient-derived xenograft (PDX) engraftment and prognosis in patients receiving up-to-date treatments for cancers classified as metastatic (stage M) high-risk neuroblastoma (HR-NB) by the International Neuroblastoma Risk Group Staging System (INRGSS). METHODS We obtained biopsies from patients with stage M HR-NB. We inoculated biopsy fragments subcutaneously in mice. We studied the association of PDX engraftment with event-free survival (EFS) and overall survival (OS) of patients. RESULTS Since 2009, we established 17 PDX from 97 samples of 66 patients with stage M HR-NB, with a follow-up of at least two years. Factors associated with higher probability of engraftment were the death as outcome (p = .0006) and the amplification of the gene MYCN in tumors (p = .0271). Patients whose biopsies established a PDX had significantly shorter EFS and OS (p = .0039 and .0002, respectively) than patients whose samples did not engraft. The association of PDX engraftment and OS was significant in patients without MYCN amplification (p = .0041), but not in patients with MYCN amplification (p = .2707). CONCLUSION Positive PDX engraftment is a factor related to poor prognosis and fatal outcome in patients with stage M HR-NB treated with up-to-date therapies.
Collapse
Affiliation(s)
- Rosario Aschero
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Helena Castillo-Ecija
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Merce Baulenas-Farres
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Claudia Resa-Pares
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Ana Jimenez-Cabaco
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Eva Rodriguez
- Department of Pathology, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Carles Monterrubio
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Sara Perez-Jaume
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Mariona Suñol
- Department of Pathology, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Guillermo L Chantada
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
- CONICET, Buenos Aires, Argentina
| | - Cinzia Lavarino
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Jaume Mora
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Angel M Carcaboso
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| |
Collapse
|
11
|
Chen A, Neuwirth I, Herndler-Brandstetter D. Modeling the Tumor Microenvironment and Cancer Immunotherapy in Next-Generation Humanized Mice. Cancers (Basel) 2023; 15:2989. [PMID: 37296949 PMCID: PMC10251926 DOI: 10.3390/cancers15112989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/10/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Cancer immunotherapy has brought significant clinical benefits to numerous patients with malignant disease. However, only a fraction of patients experiences complete and durable responses to currently available immunotherapies. This highlights the need for more effective immunotherapies, combination treatments and predictive biomarkers. The molecular properties of a tumor, intratumor heterogeneity and the tumor immune microenvironment decisively shape tumor evolution, metastasis and therapy resistance and are therefore key targets for precision cancer medicine. Humanized mice that support the engraftment of patient-derived tumors and recapitulate the human tumor immune microenvironment of patients represent a promising preclinical model to address fundamental questions in precision immuno-oncology and cancer immunotherapy. In this review, we provide an overview of next-generation humanized mouse models suitable for the establishment and study of patient-derived tumors. Furthermore, we discuss the opportunities and challenges of modeling the tumor immune microenvironment and testing a variety of immunotherapeutic approaches using human immune system mouse models.
Collapse
Affiliation(s)
| | | | - Dietmar Herndler-Brandstetter
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, 1090 Vienna, Austria; (A.C.); (I.N.)
| |
Collapse
|
12
|
Tarin M, Némati F, Decaudin D, Canbezdi C, Marande B, Silva L, Derrien H, Jochemsen AG, Gardrat S, Piperno-Neumann S, Rodrigues M, Mariani P, Cassoux N, Stern MH, Roman-Roman S, Alsafadi S. FAK Inhibitor-Based Combinations with MEK or PKC Inhibitors Trigger Synergistic Antitumor Effects in Uveal Melanoma. Cancers (Basel) 2023; 15:cancers15082280. [PMID: 37190207 DOI: 10.3390/cancers15082280] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Uveal Melanoma (UM) is a rare and malignant intraocular tumor with dismal prognosis. Even if radiation or surgery permit an efficient control of the primary tumor, up to 50% of patients subsequently develop metastases, mainly in the liver. The treatment of UM metastases is challenging and the patient survival is very poor. The most recurrent event in UM is the activation of Gαq signaling induced by mutations in GNAQ/11. These mutations activate downstream effectors including protein kinase C (PKC) and mitogen-activated protein kinases (MAPK). Clinical trials with inhibitors of these targets have not demonstrated a survival benefit for patients with UM metastasis. Recently, it has been shown that GNAQ promotes YAP activation through the focal adhesion kinase (FAK). Pharmacological inhibition of MEK and FAK showed remarkable synergistic growth-inhibitory effects in UM both in vitro and in vivo. In this study, we have evaluated the synergy of the FAK inhibitor with a series of inhibitors targeting recognized UM deregulated pathways in a panel of cell lines. The combined inhibition of FAK and MEK or PKC had highly synergistic effects by reducing cell viability and inducing apoptosis. Furthermore, we demonstrated that these combinations exert a remarkable in vivo activity in UM patient-derived xenografts. Our study confirms the previously described synergy of the dual inhibition of FAK and MEK and identifies a novel combination of drugs (FAK and PKC inhibitors) as a promising strategy for therapeutic intervention in metastatic UM.
Collapse
Affiliation(s)
- Malcy Tarin
- Translational Research Department, Institut Curie, PSL Research University, 75005 Paris, France
| | - Fariba Némati
- Laboratory of Preclinical Investigation, Institut Curie, PSL Research University, 75005 Paris, France
| | - Didier Decaudin
- Laboratory of Preclinical Investigation, Institut Curie, PSL Research University, 75005 Paris, France
- Department of Medical Oncology, Institut Curie, PSL Research University, 75005 Paris, France
| | - Christine Canbezdi
- Translational Research Department, Institut Curie, PSL Research University, 75005 Paris, France
| | - Benjamin Marande
- Translational Research Department, Institut Curie, PSL Research University, 75005 Paris, France
| | - Lisseth Silva
- Translational Research Department, Institut Curie, PSL Research University, 75005 Paris, France
| | - Héloïse Derrien
- Laboratory of Preclinical Investigation, Institut Curie, PSL Research University, 75005 Paris, France
| | - Aart G Jochemsen
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Sophie Gardrat
- Department of Biopathology, Institut Curie, PSL Research University, 75005 Paris, France
| | - Sophie Piperno-Neumann
- Department of Medical Oncology, Institut Curie, PSL Research University, 75005 Paris, France
| | - Manuel Rodrigues
- Department of Medical Oncology, Institut Curie, PSL Research University, 75005 Paris, France
- INSERM U830, DNA Repair and Uveal Melanoma, Institut Curie, PSL Research University, 75005 Paris, France
| | - Pascale Mariani
- Department of Medical Oncology, Institut Curie, PSL Research University, 75005 Paris, France
| | - Nathalie Cassoux
- Department of Ocular Oncology, Institut Curie, Université Paris Cité, 94010 Paris, France
| | - Marc-Henri Stern
- INSERM U830, DNA Repair and Uveal Melanoma, Institut Curie, PSL Research University, 75005 Paris, France
| | - Sergio Roman-Roman
- Translational Research Department, Institut Curie, PSL Research University, 75005 Paris, France
| | - Samar Alsafadi
- Translational Research Department, Institut Curie, PSL Research University, 75005 Paris, France
| |
Collapse
|
13
|
Liu Y, Wu W, Cai C, Zhang H, Shen H, Han Y. Patient-derived xenograft models in cancer therapy: technologies and applications. Signal Transduct Target Ther 2023; 8:160. [PMID: 37045827 PMCID: PMC10097874 DOI: 10.1038/s41392-023-01419-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Patient-derived xenograft (PDX) models, in which tumor tissues from patients are implanted into immunocompromised or humanized mice, have shown superiority in recapitulating the characteristics of cancer, such as the spatial structure of cancer and the intratumor heterogeneity of cancer. Moreover, PDX models retain the genomic features of patients across different stages, subtypes, and diversified treatment backgrounds. Optimized PDX engraftment procedures and modern technologies such as multi-omics and deep learning have enabled a more comprehensive depiction of the PDX molecular landscape and boosted the utilization of PDX models. These irreplaceable advantages make PDX models an ideal choice in cancer treatment studies, such as preclinical trials of novel drugs, validating novel drug combinations, screening drug-sensitive patients, and exploring drug resistance mechanisms. In this review, we gave an overview of the history of PDX models and the process of PDX model establishment. Subsequently, the review presents the strengths and weaknesses of PDX models and highlights the integration of novel technologies in PDX model research. Finally, we delineated the broad application of PDX models in chemotherapy, targeted therapy, immunotherapy, and other novel therapies.
Collapse
Affiliation(s)
- Yihan Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
| |
Collapse
|
14
|
Tsimpaki T, Bechrakis NE, Seitz B, Kraemer MM, Liu H, Dalbah S, Sokolenko E, Berchner-Pfannschmidt U, Fiorentzis M. Chick Chorioallantoic Membrane as a Patient-Derived Xenograft Model for Uveal Melanoma: Imaging Modalities for Growth and Vascular Evaluation. Cancers (Basel) 2023; 15:cancers15051436. [PMID: 36900228 PMCID: PMC10000919 DOI: 10.3390/cancers15051436] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Patient-derived tumor xenografts (PDXs) have emerged as valuable preclinical in vivo models in oncology as they largely retain the polygenomic architecture of the human tumors from which they originate. Although animal models are accompanied by cost and time constraints and a low engraftment rate, PDXs have primarily been established in immunodeficient rodent models for the in vivo assessment of tumor characteristics and of novel therapeutic cancer targets. The chick chorioallantoic membrane (CAM) assay represents an attractive alternative in vivo model that has long been used in the research of tumor biology and angiogenesis, and can overcome some of these limitations. METHODS In this study, we reviewed different technical approaches for the establishment and monitoring of a CAM-based uveal melanoma PDX model. Forty-six fresh tumor grafts were acquired after enucleation from six uveal melanoma patients and were implanted onto the CAM on ED7 with Matrigel and a ring (group 1), with Matrigel (group 2), or natively without Matrigel or a ring (group 3). Real-time imaging techniques, such as various ultrasound modalities, optical coherence tomography, infrared imaging, and imaging analyses with Image J for tumor growth and extension, as well as color doppler, optical coherence angiography, and fluorescein angiography for angiogenesis, were performed on ED18 as alternative monitoring instruments. The tumor samples were excised on ED18 for histological assessment. RESULTS There were no significant differences between the three tested experimental groups regarding the length and width of the grafts during the development period. A statistically significant increase in volume (p = 0.0007) and weight (p = 0.0216) between ED7 and ED18 was only documented for tumor specimens of group 2. A significant correlation of the results for the cross-sectional area, largest basal diameter, and volume was documented between the different imaging and measurement techniques and the excised grafts. The formation of a vascular star around the tumor and of a vascular ring on the base of the tumor was observed for the majority of the viable developing grafts as a sign of successful engraftment. CONCLUSION The establishment of a CAM-PDX uveal melanoma model could elucidate the biological growth patterns and the efficacy of new therapeutic options in vivo. The methodological novelty of this study, investigating different implanting techniques and exploiting advances in real-time imaging with multiple modalities, allows precise, quantitative assessment in the field of tumor experimentation, underlying the feasibility of CAM as an in vivo PDX model.
Collapse
Affiliation(s)
- Theodora Tsimpaki
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany
| | - Nikolaos E. Bechrakis
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany
| | - Berthold Seitz
- Department of Ophthalmology, Saarland University Medical Center, Kirrberger Str. 100, 66421 Homburg, Germany
| | - Miriam M. Kraemer
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany
| | - Hongtao Liu
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany
| | - Sami Dalbah
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany
| | - Ekaterina Sokolenko
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany
| | - Utta Berchner-Pfannschmidt
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany
| | - Miltiadis Fiorentzis
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany
- Correspondence: ; Tel.: +49-723-84378
| |
Collapse
|
15
|
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: 1.0] [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
|
16
|
Liu X, Xin Z, Wang K. Patient-derived xenograft model in colorectal cancer basic and translational research. Animal Model Exp Med 2023; 6:26-40. [PMID: 36543756 PMCID: PMC9986239 DOI: 10.1002/ame2.12299] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most popular malignancies globally, with 930 000 deaths in 2020. The evaluation of CRC-related pathogenesis and the discovery of potential therapeutic targets will be meaningful and helpful for improving CRC treatment. With huge efforts made in past decades, the systematic treatment regimens have been applied to improve the prognosis of CRC patients. However, the sensitivity of CRC to chemotherapy and targeted therapy is different from person to person, which is an important cause of treatment failure. The emergence of patient-derived xenograft (PDX) models shows great potential to alleviate the straits. PDX models possess similar genetic and pathological characteristics as the features of primary tumors. Moreover, PDX has the ability to mimic the tumor microenvironment of the original tumor. Thus, the PDX model is an important tool to screen precise drugs for individualized treatment, seek predictive biomarkers for prognosis supervision, and evaluate the unknown mechanism in basic research. This paper reviews the recent advances in constructed methods and applications of the CRC PDX model, aiming to provide new knowledge for CRC basic research and therapeutics.
Collapse
Affiliation(s)
- Xiaofeng Liu
- Hepatopancreatobiliary Surgery Department I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zechang Xin
- Hepatopancreatobiliary Surgery Department I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Kun Wang
- Hepatopancreatobiliary Surgery Department I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
17
|
Qin T, Fan J, Lu F, Zhang L, Liu C, Xiong Q, Zhao Y, Chen G, Sun C. Harnessing preclinical models for the interrogation of ovarian cancer. J Exp Clin Cancer Res 2022; 41:277. [PMID: 36114548 PMCID: PMC9479310 DOI: 10.1186/s13046-022-02486-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 09/05/2022] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer (OC) is a heterogeneous malignancy with various etiology, histopathology, and biological feature. Despite accumulating understanding of OC in the post-genomic era, the preclinical knowledge still undergoes limited translation from bench to beside, and the prognosis of ovarian cancer has remained dismal over the past 30 years. Henceforth, reliable preclinical model systems are warranted to bridge the gap between laboratory experiments and clinical practice. In this review, we discuss the status quo of ovarian cancer preclinical models which includes conventional cell line models, patient-derived xenografts (PDXs), patient-derived organoids (PDOs), patient-derived explants (PDEs), and genetically engineered mouse models (GEMMs). Each model has its own strengths and drawbacks. We focus on the potentials and challenges of using these valuable tools, either alone or in combination, to interrogate critical issues with OC.
Collapse
|
18
|
Patten LW, Blatchford P, Strand M, Kaizer AM. Assessing the performance of different outcomes for tumor growth studies with animal models. Animal Model Exp Med 2022; 5:248-257. [PMID: 35699330 PMCID: PMC9240739 DOI: 10.1002/ame2.12250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/17/2022] [Indexed: 11/22/2022] Open
Abstract
The consistency of reporting results for patient‐derived xenograft (PDX) studies is an area of concern. The PDX method commonly starts by implanting a derivative of a human tumor into a mouse, then comparing the tumor growth under different treatment conditions. Currently, a wide array of statistical methods (e.g., t‐test, regression, chi‐squared test) are used to analyze these data, which ultimately depend on the outcome chosen (e.g., tumor volume, relative growth, categorical growth). In this simulation study, we provide empirical evidence for the outcome selection process by comparing the performance of both commonly used outcomes and novel variations of common outcomes used in PDX studies. Data were simulated to mimic tumor growth under multiple scenarios, then each outcome of interest was evaluated for 10 000 iterations. Comparisons between different outcomes were made with respect to average bias, variance, type‐1 error, and power. A total of 18 continuous, categorical, and time‐to‐event outcomes were evaluated, with ultimately 2 outcomes outperforming the others: final tumor volume and change in tumor volume from baseline. Notably, the novel variations of the tumor growth inhibition index (TGII)—a commonly used outcome in PDX studies—was found to perform poorly in several scenarios with inflated type‐1 error rates and a relatively large bias. Finally, all outcomes of interest were applied to a real‐world dataset.
Collapse
Affiliation(s)
- Luke W Patten
- Department of Biostatistics and Informatics, University of Colorado, Aurora, Colorado, USA
| | - Patrick Blatchford
- Department of Biostatistics and Informatics, University of Colorado, Aurora, Colorado, USA
| | - Matthew Strand
- Department of Biostatistics and Informatics, University of Colorado, Aurora, Colorado, USA
| | - Alexander M Kaizer
- Department of Biostatistics and Informatics, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
19
|
Sabat‐Pośpiech D, Fabian‐Kolpanowicz K, Kalirai H, Kipling N, Coupland SE, Coulson JM, Fielding AB. Aggressive uveal melanoma displays a high degree of centrosome amplification, opening the door to therapeutic intervention. J Pathol Clin Res 2022; 8:383-394. [PMID: 35474453 PMCID: PMC9161346 DOI: 10.1002/cjp2.272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/18/2022] [Accepted: 03/28/2022] [Indexed: 11/22/2022]
Abstract
Uveal melanoma (UM) is the most common intraocular cancer in adults. Whilst treatment of primary UM (PUM) is often successful, around 50% of patients develop metastatic disease with poor outcomes, linked to chromosome 3 loss (monosomy 3, M3). Advances in understanding UM cell biology may indicate new therapeutic options. We report that UM exhibits centrosome abnormalities, which in other cancers are associated with increased invasiveness and worse prognosis, but also represent a potential Achilles' heel for cancer-specific therapeutics. Analysis of 75 PUM patient samples revealed both higher centrosome numbers and an increase in centrosomes with enlarged pericentriolar matrix (PCM) compared to surrounding normal tissue, both indicative of centrosome amplification. The PCM phenotype was significantly associated with M3 (t-test, p < 0.01). Centrosomes naturally enlarge as cells approach mitosis; however, whilst UM with higher mitotic scores had enlarged PCM regardless of genetic status, the PCM phenotype remained significantly associated with M3 in UM with low mitotic scores (ANOVA, p = 0.021) suggesting that this is independent of proliferation. Phenotypic analysis of patient-derived cultures and established UM lines revealed comparable levels of centrosome amplification in PUM cells to archetypal triple-negative breast cancer cell lines, whilst metastatic UM (MUM) cell lines had even higher levels. Importantly, many UM cells also exhibit centrosome clustering, a common strategy employed by other cancer cells with centrosome amplification to survive cell division. As UM samples with M3 display centrosome abnormalities indicative of amplification, this phenotype may contribute to the development of MUM, suggesting that centrosome de-clustering drugs may provide a novel therapeutic approach.
Collapse
Affiliation(s)
- Dorota Sabat‐Pośpiech
- Molecular Physiology and Cell Signalling, Institute of Systems Molecular & Integrative BiologyUniversity of LiverpoolLiverpoolUK
- Molecular and Clinical Cancer Medicine, Institute of Systems Molecular & Integrative BiologyUniversity of LiverpoolLiverpoolUK
| | - Kim Fabian‐Kolpanowicz
- Biomedical and Life Sciences, Faculty of Health and MedicineLancaster UniversityLancasterUK
| | - Helen Kalirai
- Molecular and Clinical Cancer Medicine, Institute of Systems Molecular & Integrative BiologyUniversity of LiverpoolLiverpoolUK
| | - Natalie Kipling
- Molecular and Clinical Cancer Medicine, Institute of Systems Molecular & Integrative BiologyUniversity of LiverpoolLiverpoolUK
| | - Sarah E Coupland
- Molecular and Clinical Cancer Medicine, Institute of Systems Molecular & Integrative BiologyUniversity of LiverpoolLiverpoolUK
| | - Judy M Coulson
- Molecular Physiology and Cell Signalling, Institute of Systems Molecular & Integrative BiologyUniversity of LiverpoolLiverpoolUK
| | - Andrew B Fielding
- Molecular Physiology and Cell Signalling, Institute of Systems Molecular & Integrative BiologyUniversity of LiverpoolLiverpoolUK
- Biomedical and Life Sciences, Faculty of Health and MedicineLancaster UniversityLancasterUK
| |
Collapse
|
20
|
Passeri T, Dahmani A, Masliah-Planchon J, Naguez A, Michou M, El Botty R, Vacher S, Bouarich R, Nicolas A, Polivka M, Franck C, Schnitzler A, Némati F, Roman-Roman S, Bourdeaut F, Adle-Biassette H, Mammar H, Froelich S, Bièche I, Decaudin D. Dramatic In Vivo Efficacy of the EZH2-Inhibitor Tazemetostat in PBRM1-Mutated Human Chordoma Xenograft. Cancers (Basel) 2022; 14:cancers14061486. [PMID: 35326637 PMCID: PMC8946089 DOI: 10.3390/cancers14061486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Chordomas are rare bone tumors characterized by a high recurrence rate. Presently, no medical treatment is available for advanced diseases due to the lack of molecular data and preclinical models. The current study showed the establishment and characterization of the largest panel chordoma xenografts, allowing pharmacological studies. In one PBRM1-mutated model, we demonstrated a strong therapeutic efficacy of the EZH2-inhibitor tazemetostat, encouraging further research on EZH2-inhibitors in chordomas. Abstract Chordomas are rare neoplasms characterized by a high recurrence rate and a poor long-term prognosis. Considering their chemo-/radio-resistance, alternative treatment strategies are strongly required, but their development is limited by the paucity of relevant preclinical models. Mutations affecting genes of the SWI/SNF complexes are frequently found in chordomas, suggesting a potential therapeutic effect of epigenetic regulators in this pathology. Twelve PDX models were established and characterized on histological and biomolecular features. Patients whose tumors were able to grow into mice had a statistically significant lower progression-free survival than those whose tumors did not grow after in vivo transplantation (p = 0.007). All PDXs maintained the same histopathological features as patients’ tumors. Homozygous deletions of CDKN2A/2B (58.3%) and PBRM1 (25%) variants were the most common genomic alterations found. In the tazemetostat treated PDX model harboring a PBRM1 variant, an overall survival of 100% was observed. Our panel of chordoma PDXs represents a useful preclinical tool for both pharmacologic and biological assessments. The first demonstration of a high antitumor activity of tazemetostat in a PDX model harboring a PBRM1 variant supports further evaluation for EZH2-inhibitors in this subgroup of chordomas.
Collapse
Affiliation(s)
- Thibault Passeri
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, University of Paris Saclay, 75005 Paris, France; (T.P.); (A.D.); (A.N.); (M.M.); (R.E.B.); (F.N.)
- Department of Genetics, Institut Curie, University of Paris Saclay, 75005 Paris, France; (J.M.-P.); (S.V.); (C.F.); (A.S.); (I.B.)
- Department of Neurosurgery, Lariboisière Hospital, Assistance Publique des Hôpitaux de Paris, University of Paris, 75010 Paris, France;
| | - Ahmed Dahmani
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, University of Paris Saclay, 75005 Paris, France; (T.P.); (A.D.); (A.N.); (M.M.); (R.E.B.); (F.N.)
| | - Julien Masliah-Planchon
- Department of Genetics, Institut Curie, University of Paris Saclay, 75005 Paris, France; (J.M.-P.); (S.V.); (C.F.); (A.S.); (I.B.)
| | - Adnan Naguez
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, University of Paris Saclay, 75005 Paris, France; (T.P.); (A.D.); (A.N.); (M.M.); (R.E.B.); (F.N.)
| | - Marine Michou
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, University of Paris Saclay, 75005 Paris, France; (T.P.); (A.D.); (A.N.); (M.M.); (R.E.B.); (F.N.)
| | - Rania El Botty
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, University of Paris Saclay, 75005 Paris, France; (T.P.); (A.D.); (A.N.); (M.M.); (R.E.B.); (F.N.)
| | - Sophie Vacher
- Department of Genetics, Institut Curie, University of Paris Saclay, 75005 Paris, France; (J.M.-P.); (S.V.); (C.F.); (A.S.); (I.B.)
| | - Rachida Bouarich
- Integrated Cancer Research Site, Institut Curie, 75005 Paris, France; (R.B.); (F.B.)
| | - André Nicolas
- Department of Tumor Biology, Institut Curie, 75005 Paris, France;
| | - Marc Polivka
- Department of Pathology, Lariboisière Hospital, Assistance Publique des Hôpitaux de Paris, University of Paris, UMR 1141 Inserm, 75010 Paris, France; (M.P.); (H.A.-B.)
| | - Coralie Franck
- Department of Genetics, Institut Curie, University of Paris Saclay, 75005 Paris, France; (J.M.-P.); (S.V.); (C.F.); (A.S.); (I.B.)
| | - Anne Schnitzler
- Department of Genetics, Institut Curie, University of Paris Saclay, 75005 Paris, France; (J.M.-P.); (S.V.); (C.F.); (A.S.); (I.B.)
| | - Fariba Némati
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, University of Paris Saclay, 75005 Paris, France; (T.P.); (A.D.); (A.N.); (M.M.); (R.E.B.); (F.N.)
| | - Sergio Roman-Roman
- Department of Translational Research, Institut Curie, University of Paris Saclay, 75005 Paris, France;
| | - Franck Bourdeaut
- Integrated Cancer Research Site, Institut Curie, 75005 Paris, France; (R.B.); (F.B.)
| | - Homa Adle-Biassette
- Department of Pathology, Lariboisière Hospital, Assistance Publique des Hôpitaux de Paris, University of Paris, UMR 1141 Inserm, 75010 Paris, France; (M.P.); (H.A.-B.)
| | - Hamid Mammar
- Proton Therapy Center, Institut Curie, 91400 Orsay, France;
| | - Sébastien Froelich
- Department of Neurosurgery, Lariboisière Hospital, Assistance Publique des Hôpitaux de Paris, University of Paris, 75010 Paris, France;
| | - Ivan Bièche
- Department of Genetics, Institut Curie, University of Paris Saclay, 75005 Paris, France; (J.M.-P.); (S.V.); (C.F.); (A.S.); (I.B.)
| | - Didier Decaudin
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, University of Paris Saclay, 75005 Paris, France; (T.P.); (A.D.); (A.N.); (M.M.); (R.E.B.); (F.N.)
- Department of Medical Oncology, Institut Curie, 75005 Paris, France
- Correspondence: ; Tel.: +33-1-56-24-62-40
| |
Collapse
|
21
|
Beserra AO, Estevan EC, Bezerra SM, Torrezan GT, Ikegami A, Dellê H, Cunha IW, Meira IT, Carraro DM, Lara PN, Zequi SC, Martins VR, Santos TG. Patient-Derived Renal Cell Carcinoma Xenografts Capture Tumor Genetic Profiles and Aggressive Behaviors. KIDNEY CANCER 2022. [DOI: 10.3233/kca-210011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Patient-derived xenografts (PDX) have emerged as one of the most promising model systems to study cancer biology and to develop new antineoplastic drugs. Renal cell carcinoma (RCC) represents up to 90% of all kidney tumors, exhibits aggressive behavior, and has a propensity for metastasis. At diagnosis, 30% of patients with RCC have metastases, while up to 50% of those with localized disease treated with curative protocols experience recurrence. OBJECTIVE: This study aimed to establish an RCC PDX platform to identify novel clinical and molecular biomarkers of recurrence risk in order to facilitate precision medicine. METHODS: Tumor samples were obtained from surgical specimens of 87 RCC patients; fragments were implanted in immunodeficient NOD/SCID/gamma (NSG) mice. Seventeen fragments were implanted subcutaneously in an initial group while a second group of 70 samples were implanted orthotopically in the subcapsular space. RESULTS: A total of 19 PDX developed only after orthotopic implantation, and included 15 cases of clear cell RCC subtype, 3 cases of papillary subtype, and one unclassifiable tumor. One PDX of clear cell RCC recapitulated the phenotype of vena caval tumor thrombus extension that had been diagnosed in the source patient. PDX characterization by immunohistochemistry and targeted sequencing indicated that all PDXs preserved RCC identity and major molecular alterations. Moreover, the capacity of tumor engraftment was a strong prognostic indicator for patients with locally advanced disease. CONCLUSION: Taken together, these results suggest that the orthotopic xenograft model of RCC represents a suitable tool to study RCC biology, identify biomarkers, and to test therapeutic candidates.
Collapse
Affiliation(s)
- Adriano O. Beserra
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| | - Ethiene C. Estevan
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| | | | - Giovana T. Torrezan
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| | - Amanda Ikegami
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| | - Humberto Dellê
- Graduate Program in Medicine, Universidade Nove de Julho, São Paulo – Brazil
| | - Isabela W. Cunha
- Institute of Pathology, Rede D’OR-São Luiz and D’Or Institute for Research and Education (IDOR), São Paulo – Brazil
| | - Isabella T. Meira
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| | - Dirce M. Carraro
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| | - Primo N. Lara
- University of California Davis Comprehensive Cancer Center, Sacramento, CA – USA
| | - Stenio C. Zequi
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
- Reference Center of Urology, A.C. Camargo Cancer Center, São Paulo – Brazil
- LARCG -Latin American Renal Cancer Group
| | - Vilma R. Martins
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| | - Tiago G. Santos
- International Research Center, A.C. Camargo Cancer Center, São Paulo – Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo – Brazil
| |
Collapse
|
22
|
Letai A, Bhola P, Welm AL. Functional precision oncology: Testing tumors with drugs to identify vulnerabilities and novel combinations. Cancer Cell 2022; 40:26-35. [PMID: 34951956 PMCID: PMC8752507 DOI: 10.1016/j.ccell.2021.12.004] [Citation(s) in RCA: 114] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/26/2021] [Accepted: 12/02/2021] [Indexed: 01/12/2023]
Abstract
Functional precision medicine is a strategy whereby live tumor cells from affected individuals are directly perturbed with drugs to provide immediately translatable, personalized information to guide therapy. The heterogeneity of human cancer has led to the realization that personalized approaches are needed to improve treatment outcomes. Precision oncology has traditionally used static features of the tumor to dictate which therapies should be used. Static features can include expression of key targets or genomic analysis of mutations to identify therapeutically targetable "drivers." Although a surprisingly small proportion of individuals derive clinical benefit from the static approach, functional precision medicine can provide additional information regarding tumor vulnerabilities. We discuss emerging technologies for functional precision medicine as well as limitations and challenges in using these assays in the clinical trials that will be necessary to determine whether functional precision medicine can improve outcomes and eventually become a standard tool in clinical oncology.
Collapse
Affiliation(s)
- Anthony Letai
- Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02215, USA
| | - Patrick Bhola
- Harvard Medical School, Boston, MA 02215, USA; Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Alana L Welm
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
23
|
Evaluation of two in vitro assays for tumorigenicity assessment of CRISPR-Cas9 genome-edited cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 23:241-253. [PMID: 34703845 PMCID: PMC8505356 DOI: 10.1016/j.omtm.2021.09.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 09/03/2021] [Indexed: 12/26/2022]
Abstract
Off-target editing is one of the main safety concerns for the use of CRISPR-Cas9 genome editing in gene therapy. These unwanted modifications could lead to malignant transformation, which renders tumorigenicity assessment of gene therapy products indispensable. In this study, we established two in vitro transformation assays, the soft agar colony-forming assay (SACF) and the growth in low attachment assay (GILA) as alternative methods for tumorigenicity evaluation of genome-edited cells. Using a CRISPR-Cas9-based approach to transform immortalized MCF10A cells, we identified PTPN12, a known tumor suppressor, as a valid positive control in GILA and SACF. Next, we measured the limit of detection for both assays and proved that SACF is more sensitive than GILA (0.8% versus 3.1% transformed cells). We further validated SACF and GILA by identifying a set of positive and negative controls and by testing the suitability of another cell line (THLE-2). Moreover, in contrast to SACF and GILA, an in vivo tumorigenicity study failed to detect the known tumorigenic potential of PTPN12 deletion, demonstrating the relevance of GILA and SACF in tumorigenicity testing. In conclusion, SACF and GILA are both attractive and valuable additions to preclinical safety assessment of gene therapy products.
Collapse
|
24
|
Long JE, Jankovic M, Maddalo D. Drug discovery oncology in a mouse: concepts, models and limitations. Future Sci OA 2021; 7:FSO737. [PMID: 34295539 PMCID: PMC8288236 DOI: 10.2144/fsoa-2021-0019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/27/2021] [Indexed: 02/08/2023] Open
Abstract
The utilization of suitable mouse models is a critical step in the drug discovery oncology workflow as their generation and use are important for target identification and validation as well as toxicity and efficacy assessments. Current murine models have been instrumental in furthering insights into the mode of action of drugs before transitioning into the clinic. Recent advancements in genome editing with the development of the CRISPR/Cas9 system and the possibility of applying such technology directly in vivo have expanded the toolkit of preclinical models available. In this review, a brief presentation of the current models used in drug discovery will be provided with a particular emphasis on the novel CRISPR/Cas9 models.
Collapse
Affiliation(s)
- Jason E Long
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Maja Jankovic
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, QC, H4A 3J1, Canada
- Lady Davis Institute for Medical Research, Montréal, QC, H4A 3J1, Canada
| | - Danilo Maddalo
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA 94080, USA
- Pharmaceutical Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, 4070, Switzerland
- Author for correspondence:
| |
Collapse
|
25
|
Piyawajanusorn C, Nguyen LC, Ghislat G, Ballester PJ. A gentle introduction to understanding preclinical data for cancer pharmaco-omic modeling. Brief Bioinform 2021; 22:6343527. [PMID: 34368843 DOI: 10.1093/bib/bbab312] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/25/2021] [Accepted: 07/20/2021] [Indexed: 12/16/2022] Open
Abstract
A central goal of precision oncology is to administer an optimal drug treatment to each cancer patient. A common preclinical approach to tackle this problem has been to characterize the tumors of patients at the molecular and drug response levels, and employ the resulting datasets for predictive in silico modeling (mostly using machine learning). Understanding how and why the different variants of these datasets are generated is an important component of this process. This review focuses on providing such introduction aimed at scientists with little previous exposure to this research area.
Collapse
Affiliation(s)
- Chayanit Piyawajanusorn
- Cancer Research Center of Marseille, INSERM U1068, F-13009 Marseille, France.,Institut Paoli-Calmettes, F-13009 Marseille, France.,Aix-Marseille Université, F-13284 Marseille, France.,CNRS UMR7258, F-13009 Marseille, France.,Faculty of Medicine and Public Health, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Linh C Nguyen
- Cancer Research Center of Marseille, INSERM U1068, F-13009 Marseille, France.,Institut Paoli-Calmettes, F-13009 Marseille, France.,Aix-Marseille Université, F-13284 Marseille, France.,CNRS UMR7258, F-13009 Marseille, France.,Department of Life Sciences, University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Ghita Ghislat
- U1104, CNRS UMR7280, Centre d'Immunologie de Marseille-Luminy, Inserm, Marseille, France
| | - Pedro J Ballester
- Cancer Research Center of Marseille, INSERM U1068, F-13009 Marseille, France.,Institut Paoli-Calmettes, F-13009 Marseille, France.,Aix-Marseille Université, F-13284 Marseille, France.,CNRS UMR7258, F-13009 Marseille, France
| |
Collapse
|
26
|
Terai M, Kageyama K, Sugase T, Lam BQ, Alexeev V, Sato T. Orthotopic Human Metastatic Uveal Melanoma Xenograft Mouse Models: Applications for Understanding the Pathophysiology and Therapeutic Management of Metastatic Uveal Melanoma. Curr Protoc 2021; 1:e110. [PMID: 33882197 DOI: 10.1002/cpz1.110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The propensity of uveal melanoma to metastasize to the liver hinders the accrual of micro-metastatic and end-stage disease tissue samples and restricts the investigation of metastatic uveal melanoma (MUM). Pre-clinical experimental animal models of MUM can help elucidate the pathophysiology of metastatic lesions and provide a tool for designing new therapeutic approaches for MUM. Here, we present an advanced model of hepatic metastases that enables quantitatively visualizing the development of individual hepatic tumor clones and estimating their growth kinetics and colonization efficiency. Similar to clinically observed liver metastases, these models enable the assessment of growth kinetics of the liver micro-metastases and the testing of therapeutic approaches for the treatment of MUM. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Experimental patient-derived xenograft mouse model of metastatic uveal melanoma Basic Protocol 2: Experimental liver micro-metastatic mouse model using splenic injection of metastatic uveal melanoma cells.
Collapse
Affiliation(s)
- Mizue Terai
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ken Kageyama
- Department of Diagnostic and Interventional Radiology, Osaka City University, Osaka-shi, Osaka, Japan
| | - Takahito Sugase
- Department of Gastrointestinal Surgery, Graduate School of Medicine, Osaka University, Suita-shi, Osaka, Japan
| | - Bao Quoc Lam
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Vitali Alexeev
- Department of Dermatology & Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Takami Sato
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
27
|
Goldrick C, Palanga L, Tang B, Mealy G, Crown J, Horgan N, Kennedy S, Walsh N. Hindsight: Review of Preclinical Disease Models for the Development of New Treatments for Uveal Melanoma. J Cancer 2021; 12:4672-4685. [PMID: 34149931 PMCID: PMC8210544 DOI: 10.7150/jca.53954] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 05/19/2021] [Indexed: 01/01/2023] Open
Abstract
The molecular, histopathological, genomic and transcriptomic characteristics of uveal melanoma (UM) have identified four molecular subgroups with different clinical outcomes. Despite the improvements in UM classification and biological pathology, current treatments do not reduce the occurrence of metastasis. The development of effective adjuvant and metastatic therapies for UM has been slow and extremely limited. Preclinical models that closely resemble the molecular and genetic UM subgroups are essential for translating molecular findings into improved clinical treatment. In this review, we provide a retrospective view of the existing preclinical models used to study UM, and give an overview of their strengths and limitations. We review targeted therapy clinical trial data to evaluate the gap in the translation of preclinical findings to human studies. Reflecting on the current high attrition rates of clinical trials for UM, preclinical models that effectively recapitulate the human in vivo situation and/or accurately reflect the subtype classifications would enhance the translational impact of experimental data and have crucial implications for the advancement of personalised medicine.
Collapse
Affiliation(s)
- Caoimhe Goldrick
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Letizia Palanga
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Bobby Tang
- Royal Victoria Eye and Ear Research Foundation, Royal Victoria Eye and Ear Hospital, Adelaide Road, Dublin, Ireland
| | - Grace Mealy
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - John Crown
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin, Ireland
- Department of Medical Oncology, St. Vincent's University Hospital, Dublin, Ireland
| | - Noel Horgan
- Royal Victoria Eye and Ear Research Foundation, Royal Victoria Eye and Ear Hospital, Adelaide Road, Dublin, Ireland
| | - Susan Kennedy
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin, Ireland
- Royal Victoria Eye and Ear Research Foundation, Royal Victoria Eye and Ear Hospital, Adelaide Road, Dublin, Ireland
| | - Naomi Walsh
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin, Ireland
| |
Collapse
|
28
|
Boughey JC, Suman VJ, Yu J, Santo K, Sinnwell JP, Carter JM, Kalari KR, Tang X, McLaughlin SA, Moreno-Aspitia A, Northfelt DW, Gray RJ, Hunt KN, Conners AL, Ingle JN, Moyer A, Weinshilboum R, Copland JA, Wang L, Goetz MP. Patient-Derived Xenograft Engraftment and Breast Cancer Outcomes in a Prospective Neoadjuvant Study (BEAUTY). Clin Cancer Res 2021; 27:4696-4699. [PMID: 34078650 DOI: 10.1158/1078-0432.ccr-21-0641] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/02/2021] [Accepted: 05/24/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Patient-derived xenografts (PDX) are a research tool for studying cancer biology and drug response phenotypes. While engraftment rates are higher for tumors with more aggressive characteristics, it is uncertain whether engraftment is prognostic for cancer recurrence. PATIENTS AND METHODS In a prospective study of patients with breast cancer treated with neoadjuvant chemotherapy (NAC) with taxane ± trastuzumab followed by anthracycline-based chemotherapy, we report the association between breast cancer events and PDX engraftment using tumors derived from treatment naïve (pre-NAC biopsies from 113 patients) and treatment resistant (post-NAC at surgery from 34 patients). Gray test was used to assess whether the cumulative incidence of a breast cancer event differs with respect to either pre-NAC PDX engraftment or post-NAC PDX engraftment. RESULTS With a median follow-up of 5.7 years, the cumulative incidence of breast cancer relapse did not differ significantly according to pre-NAC PDX engraftment (5-year rate: 13.6% vs. 13.4%; P = 0.89). However, the incidence of a breast event was greater for patients with post-NAC PDX engraftment (5-year rate: 50.0% vs. 19.6%), but this did not achieve significance (P = 0.11). CONCLUSIONS In treatment-naïve breast cancer receiving standard NAC, PDX engraftment was not prognostic for breast cancer recurrence. Further study is needed to establish whether PDX engraftment in the treatment-resistant setting is prognostic for cancer recurrence.
Collapse
Affiliation(s)
- Judy C Boughey
- Department of Surgery, Mayo Clinic, Rochester, Minnesota.
| | - Vera J Suman
- Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Jia Yu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Katelyn Santo
- Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | - Jodi M Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | - Xiaojia Tang
- Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | - Alvaro Moreno-Aspitia
- Department of Medicine (Division of Hematology/Oncology), Mayo Clinic, Jacksonville, Florida
| | - Donald W Northfelt
- Department of Medicine (Division of Hematology/Oncology), Mayo Clinic, Scottsdale, Arizona
| | | | - Katie N Hunt
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Ann Moyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Richard Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - John A Copland
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
29
|
Rizzo G, Bertotti A, Leto SM, Vetrano S. Patient-derived tumor models: a more suitable tool for pre-clinical studies in colorectal cancer. J Exp Clin Cancer Res 2021; 40:178. [PMID: 34074330 PMCID: PMC8168319 DOI: 10.1186/s13046-021-01970-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/02/2021] [Indexed: 12/15/2022] Open
Abstract
Colorectal cancer (CRC), despite the advances in screening and surveillance, remains the second most common cause of cancer death worldwide. The biological inadequacy of pre-clinical models to fully recapitulate the multifactorial etiology and the complexity of tumor microenvironment and human CRC's genetic heterogeneity has limited cancer treatment development. This has led to the development of Patient-derived models able to phenocopy as much as possible the original inter- and intra-tumor heterogeneity of CRC, reflecting the tumor microenvironment's cellular interactions. Implantation of patient tissue into immunodeficient mice hosts and the culture of tumor organoids have allowed advances in cancer biology and metastasis. This review highlights the advantages and limits of Patient-derived models as innovative and valuable pre-clinical tools to study progression and metastasis of CRC, develop novel therapeutic strategies by creating a drug screening platform, and predict the efficacy of clinical response to therapy.
Collapse
Affiliation(s)
- Giulia Rizzo
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, 20090, Milan, Italy
| | - Andrea Bertotti
- Laboratory of Translational Cancer Medicine, Candiolo Cancer Institute - FPO IRCCs, Candiolo, 10060, Torino, Italy
- Department of Oncology, University of Torino School of Medicine, Candiolo, 10060, Torino, Italy
| | - Simonetta Maria Leto
- Laboratory of Translational Cancer Medicine, Candiolo Cancer Institute - FPO IRCCs, Candiolo, 10060, Torino, Italy
| | - Stefania Vetrano
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, 20090, Milan, Italy.
- IBD Center, Department of Gastroenterology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy.
| |
Collapse
|
30
|
Dai W, Liu S, Wang S, Zhao L, Yang X, Zhou J, Wang Y, Zhang J, Zhang P, Ding K, Li Y, Pan J. Activation of transmembrane receptor tyrosine kinase DDR1-STAT3 cascade by extracellular matrix remodeling promotes liver metastatic colonization in uveal melanoma. Signal Transduct Target Ther 2021; 6:176. [PMID: 33976105 PMCID: PMC8113510 DOI: 10.1038/s41392-021-00563-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 02/14/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
Colonization is believed a rate-limiting step of metastasis cascade. However, its underlying mechanism is not well understood. Uveal melanoma (UM), which is featured with single organ liver metastasis, may provide a simplified model for realizing the complicated colonization process. Because DDR1 was identified to be overexpressed in UM cell lines and specimens, and abundant pathological deposition of extracellular matrix collagen, a type of DDR1 ligand, was noted in the microenvironment of liver in metastatic patients with UM, we postulated the hypothesis that DDR1 and its ligand might ignite the interaction between UM cells and their surrounding niche of liver thereby conferring strengthened survival, proliferation, stemness and eventually promoting metastatic colonization in liver. We tested this hypothesis and found that DDR1 promoted these malignant cellular phenotypes and facilitated metastatic colonization of UM in liver. Mechanistically, UM cells secreted TGF-β1 which induced quiescent hepatic stellate cells (qHSCs) into activated HSCs (aHSCs) which secreted collagen type I. Such a remodeling of extracellular matrix, in turn, activated DDR1, strengthening survival through upregulating STAT3-dependent Mcl-1 expression, enhancing stemness via upregulating STAT3-dependent SOX2, and promoting clonogenicity in cancer cells. Targeting DDR1 by using 7rh, a specific inhibitor, repressed proliferation and survival in vitro and in vivo outgrowth. More importantly, targeting cancer cells by pharmacological inactivation of DDR1 or targeting microenvironmental TGF-β1-collagen I loop exhibited a prominent anti-metastasis effect in mice. In conclusion, targeting DDR1 signaling and TGF-β signaling may be a novel approach to diminish hepatic metastasis in UM.
Collapse
Affiliation(s)
- Wei Dai
- Jinan University Institute of Tumor Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China.,Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| | - Shenglan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shubo Wang
- Jinan University Institute of Tumor Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Li Zhao
- Jinan University Institute of Tumor Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Xiao Yang
- Jinan University Institute of Tumor Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jingfeng Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yun Wang
- Jinan University Institute of Tumor Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jing Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ping Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yangqiu Li
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
31
|
Prat M, Salon M, Allain T, Dubreuil O, Noël G, Preisser L, Jean B, Cassard L, Lemée F, Tabah-Fish I, Pipy B, Jeannin P, Prost JF, Barret JM, Coste A. Murlentamab, a Low Fucosylated Anti-Müllerian Hormone Type II Receptor (AMHRII) Antibody, Exhibits Anti-Tumor Activity through Tumor-Associated Macrophage Reprogrammation and T Cell Activation. Cancers (Basel) 2021; 13:cancers13081845. [PMID: 33924378 PMCID: PMC8070390 DOI: 10.3390/cancers13081845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/19/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary AMHRII, the anti-Müllerian hormone receptor, is selectively expressed in normal sexual organs in healthy adults but is also re-expressed in ovarian, colorectal and lung cancers. In this context, we developed murlentamab, a humanized glyco-engineered anti-AMHRII monoclonal antibody, currently in clinical trial. Preliminary data suggest that murlentamab anti-tumor activity involves immune response activation. Thus, in vitro experiments were performed to precisely characterize the murlentamab effect on the human immune system. We show that murlentamab treatment is associated with evidences of innate and adaptive immune cell activation in cancer patient samples. Moreover, we demonstrate that the murlentamab opsonization of AMHRII-expressing ovarian tumor cells promotes a polarization switch of both naïve and tumor-associated macrophages towards an anti-tumor M1-like phenotype. Our work also supports that, through macrophage reeducation, murlentamab activates an anti-tumor adaptive immune response. Finally, the combination of murlentamab with pembrolizumab confirmed novel clinical perspectives of murlentamab association with checkpoint inhibitors and other immuno-modulators. Abstract AMHRII, the anti-Müllerian hormone receptor, is selectively expressed in normal sexual organs but is also re-expressed in gynecologic cancers. Hence, we developed murlentamab, a humanized glyco-engineered anti-AMHRII monoclonal antibody currently in clinical trial. Low-fucosylated antibodies are known to increase the antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) potency of effector cells, but some preliminary results suggest a more global murlentamab-dependent activation of the immune system. In this context, we demonstrate here that the murlentamab opsonization of AMHRII-expressing ovarian tumor cells, in the presence of unstimulated- or tumor-associated macrophage (TAM)-like macrophages, significantly promotes macrophage-mediated ADCC and shifts the whole microenvironment towards a pro-inflammatory and anti-tumoral status, thus triggering anti-tumor activity. We also report that murlentamab orients both unstimulated- and TAM-like macrophages to an M1-like phenotype characterized by a strong expression of co-stimulation markers, pro-inflammatory cytokines and chemokines, favoring T cell recruitment and activation. Moreover, we show that murlentamab treatment shifts CD4+ Th1/Th2 balance towards a Th1 response and activates CD8+ T cells. Altogether, these results suggest that murlentamab, through naïve macrophage orientation and TAM reprogrammation, stimulates the anti-tumor adaptive immune response. Those mechanisms might contribute to the sustained clinical benefit observed in advanced cancer patients treated with murlentamab. Finally, the enhanced murlentamab activity in combination with pembrolizumab opens new therapeutic perspectives.
Collapse
Affiliation(s)
- Mélissa Prat
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
| | - Marie Salon
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, UPS, 31100 Toulouse, France
| | - Thibault Allain
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
| | - Olivier Dubreuil
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Grégory Noël
- Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium;
| | - Laurence Preisser
- Univ Angers, Université de Nantes, CHU Angers, Inserm, CRCINA, SFR ICAT, 49000 Angers, France; (L.P.); (P.J.)
| | - Bérangère Jean
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Lydie Cassard
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy, 94905 Villejuif, France;
| | - Fanny Lemée
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Isabelle Tabah-Fish
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Bernard Pipy
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, UPS, 31100 Toulouse, France
| | - Pascale Jeannin
- Univ Angers, Université de Nantes, CHU Angers, Inserm, CRCINA, SFR ICAT, 49000 Angers, France; (L.P.); (P.J.)
| | - Jean-François Prost
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Jean-Marc Barret
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Agnès Coste
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, UPS, 31100 Toulouse, France
- Correspondence: ; Tel.: +33-534-609-501
| |
Collapse
|
32
|
Bigot J, Lalanne AI, Lucibello F, Gueguen P, Houy A, Dayot S, Ganier O, Gilet J, Tosello J, Nemati F, Pierron G, Waterfall JJ, Barnhill R, Gardrat S, Piperno-Neumann S, Popova T, Masson V, Loew D, Mariani P, Cassoux N, Amigorena S, Rodrigues M, Alsafadi S, Stern MH, Lantz O. Splicing Patterns in SF3B1-Mutated Uveal Melanoma Generate Shared Immunogenic Tumor-Specific Neoepitopes. Cancer Discov 2021; 11:1938-1951. [PMID: 33811047 DOI: 10.1158/2159-8290.cd-20-0555] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 01/29/2021] [Accepted: 03/31/2021] [Indexed: 11/16/2022]
Abstract
Disruption of splicing patterns due to mutations of genes coding splicing factors in tumors represents a potential source of tumor neoantigens, which would be both public (shared between patients) and tumor-specific (not expressed in normal tissues). In this study, we show that mutations of the splicing factor SF3B1 in uveal melanoma generate such immunogenic neoantigens. Memory CD8+ T cells specific for these neoantigens are preferentially found in 20% of patients with uveal melanoma bearing SF3B1-mutated tumors. Single-cell analyses of neoepitope-specific T cells from the blood identified large clonal T-cell expansions, with distinct effector transcription patterns. Some of these expanded T-cell receptors are also present in the corresponding tumors. CD8+ T-cell clones specific for the neoepitopes specifically recognize and kill SF3B1-mutated tumor cells, supporting the use of this new family of neoantigens as therapeutic targets. SIGNIFICANCE: Mutations of the splicing factor SF3B1 in uveal melanoma generate shared neoantigens that are uniquely expressed by tumor cells, leading to recognition and killing by specific CD8 T cells. Mutations in splicing factors can be sources of new therapeutic strategies applicable to diverse tumors.This article is highlighted in the In This Issue feature, p. 1861.
Collapse
Affiliation(s)
- Jeremy Bigot
- INSERM U932, PSL University, Institut Curie, Paris, France
| | - Ana I Lalanne
- Laboratoire d'immunologie clinique, Institut Curie, Paris, France.,Centre d'investigation Clinique en Biothérapie, Institut Curie (CIC-BT1428), Paris, France
| | | | - Paul Gueguen
- INSERM U932, PSL University, Institut Curie, Paris, France
| | - Alexandre Houy
- INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, PSL University, Institut Curie, Paris, France
| | - Stephane Dayot
- INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, PSL University, Institut Curie, Paris, France
| | - Olivier Ganier
- INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, PSL University, Institut Curie, Paris, France
| | - Jules Gilet
- INSERM U932, PSL University, Institut Curie, Paris, France
| | - Jimena Tosello
- INSERM U932, PSL University, Institut Curie, Paris, France
| | - Fariba Nemati
- Centre d'investigation Clinique en Biothérapie, Institut Curie (CIC-BT1428), Paris, France.,Laboratory of Preclinical Investigation, Translational Research Department, PSL Research University, Institut Curie, Paris, France
| | | | - Joshua J Waterfall
- INSERM U830, PSL University, Institut Curie, Paris, France, and Department of Translational Research, PSL University, Institut Curie, Paris, France
| | - Raymond Barnhill
- Departments of Pathology and Translational Research, Institut Curie, Paris, France
| | - Sophie Gardrat
- INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, PSL University, Institut Curie, Paris, France.,Departments of Pathology and Translational Research, Institut Curie, Paris, France
| | | | - Tatiana Popova
- INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, PSL University, Institut Curie, Paris, France
| | - Vanessa Masson
- Laboratoire de Spectrométrie de Masse Protéomique, PSL University, Institut Curie, Paris, France
| | - Damarys Loew
- Laboratoire de Spectrométrie de Masse Protéomique, PSL University, Institut Curie, Paris, France
| | - Pascale Mariani
- Department of Surgical Oncology, University of Paris, Institut Curie, Paris, France
| | - Nathalie Cassoux
- Department of Surgical Oncology, University of Paris, Institut Curie, Paris, France
| | | | - Manuel Rodrigues
- INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, PSL University, Institut Curie, Paris, France.,Department of Medical Oncology, Institut Curie, Paris, France
| | - Samar Alsafadi
- INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, PSL University, Institut Curie, Paris, France.,Laboratory of Uveal Biology, Translational Research Department, Institut Curie, Paris, France
| | - Marc-Henri Stern
- INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, PSL University, Institut Curie, Paris, France
| | - Olivier Lantz
- INSERM U932, PSL University, Institut Curie, Paris, France. .,Laboratoire d'immunologie clinique, Institut Curie, Paris, France.,Centre d'investigation Clinique en Biothérapie, Institut Curie (CIC-BT1428), Paris, France
| |
Collapse
|
33
|
Kuznetsoff JN, Owens DA, Lopez A, Rodriguez DA, Chee NT, Kurtenbach S, Bilbao D, Roberts ER, Volmar CH, Wahlestedt C, Brothers SP, Harbour JW. Dual Screen for Efficacy and Toxicity Identifies HDAC Inhibitor with Distinctive Activity Spectrum for BAP1-Mutant Uveal Melanoma. Mol Cancer Res 2020; 19:215-222. [PMID: 33077485 DOI: 10.1158/1541-7786.mcr-20-0434] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/27/2020] [Accepted: 10/13/2020] [Indexed: 11/16/2022]
Abstract
Drug screens leading to successful targeted therapies in cancer have been mainly based on cell viability assays identifying inhibitors of dominantly acting oncogenes. In contrast, there has been little success in discovering targeted therapies that reverse the effects of inactivating mutations in tumor-suppressor genes. BAP1 is one such tumor suppressor that is frequently inactivated in a variety of cancers, including uveal melanoma, renal cell carcinoma, and mesothelioma. Because BAP1 is an epigenetic transcriptional regulator of developmental genes, we designed a two-phase drug screen involving a cell-based rescue screen of transcriptional repression caused by BAP1 loss, followed by an in vivo screen of lead compounds for rescue of a BAP1-deficient phenotype with minimal toxicity in Xenopus embryos. The first screen identified 9 compounds, 8 of which were HDAC inhibitors. The second screen eliminated all except one compound due to inefficacy or toxicity. The resulting lead compound, quisinostat, has a distinctive activity spectrum, including high potency against HDAC4, which was recently shown to be a key target of BAP1. Quisinostat was further validated in a mouse model and found to prevent the growth of BAP1-mutant uveal melanomas. This innovative strategy demonstrates the potential for identifying therapeutic compounds that target tumor-suppressor mutations in cancer. IMPLICATIONS: Few drugs have been identified that target mutations in tumor suppressors. Using a novel 2-step screening approach, strategy, we identified quisinostat as a candidate for therapy in BAP1-mutant uveal melanoma. HDAC4 is implicated as a key target in uveal melanoma and perhaps other BAP1-mutant cancers.
Collapse
Affiliation(s)
- Jeffim N Kuznetsoff
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Dawn A Owens
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Andy Lopez
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Daniel A Rodriguez
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Nancy T Chee
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, Florida
| | - Stefan Kurtenbach
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Daniel Bilbao
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Evan R Roberts
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Claude-Henry Volmar
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, Florida
| | - Claes Wahlestedt
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, Florida
| | - Shaun P Brothers
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, Florida
| | - J William Harbour
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
34
|
Tian H, Lyu Y, Yang YG, Hu Z. Humanized Rodent Models for Cancer Research. Front Oncol 2020; 10:1696. [PMID: 33042811 PMCID: PMC7518015 DOI: 10.3389/fonc.2020.01696] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/30/2020] [Indexed: 12/18/2022] Open
Abstract
As one of the most popular laboratory animal models, rodents have been playing crucial roles in mechanistic investigations of oncogenesis as well as anticancer drug or regimen discoveries. However, rodent tumors show different or no responses to therapies against human cancers, and thus, in recent years, increased attention has been given to mouse models with xenografted or spontaneous human cancer cells. By combining with the human immune system (HIS) mice, these models have become more sophisticated and robust, enabling in vivo exploration of human cancer immunology and immunotherapy. In this review, we summarize the pros and cons of these humanized mouse models, with a focus on their potential as an in vivo platform for human cancer research. We also discuss the strategies for further improving these models.
Collapse
Affiliation(s)
- Huimin Tian
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Yanan Lyu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China.,International Center of Future Science, Jilin University, Changchun, China
| | - Zheng Hu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| |
Collapse
|
35
|
Aughton K, Shahidipour H, Djirackor L, Coupland SE, Kalirai H. Characterization of Uveal Melanoma Cell Lines and Primary Tumor Samples in 3D Culture. Transl Vis Sci Technol 2020; 9:39. [PMID: 32832244 PMCID: PMC7414609 DOI: 10.1167/tvst.9.7.39] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023] Open
Abstract
Purpose Uveal melanoma (UM) typically spreads to the liver, where it is incurable, as there are limited therapeutic interventions available. This study aimed to standardize laboratory methods for generating three-dimensional (3D) spheroids using UM cell lines and primary UM (PUM) samples for use in drug screening. Methods Six UM cell lines and nine PUM, of differing genetic characteristics were cultured in two dimensions (2D) and three dimensions. 3D spheroid formation and growth were time monitored, and ImageJ software was used to calculate cross-sectional areas. PUM spheroids underwent immunohistochemistry for melanoma markers, nuclear BAP1, and cell proliferation. Chromosomal alterations in patient UM biopsies were compared with the corresponding 3D spheroid. In vitro drug assays testing doxorubicin and selumetinib assessed drug penetration and toxicity after 48 hours using imaging and the CellTiter-Glo 3D Cell Viability Assay. Results All six UM cell lines formed spheroids of varying sizes and compactness; six of the nine PUM samples (67%) also formed spheroids, composed of MelanA+ proliferating melanocytes and admixed macrophages. PUM spheroids were genetically identical to the original sampled tumor. In vitro drug assays showed varying penetrations into UM cell line spheroids, with doxorubicin passing into the spheroid core and selumetinib having an effect largely on peripheral cells. Both drugs caused a dose-dependent reduction in viability of 3D spheroid cells. Conclusions UM cell lines and PUM samples can successfully generate uniform 3D spheroids. PUM spheroids retain histological and genetic characteristics of the primary tumor. 3D spheroids are an important system for use in future high-throughput drug testing. Translational Relevance The use of 3D spheroids allows early-phase drug screening and is an important first step toward treatment personalization for UM patients.
Collapse
Affiliation(s)
- Karen Aughton
- Liverpool Ocular Oncology Research Group, University of Liverpool, Liverpool, United Kingdom
| | - Haleh Shahidipour
- Translational Health Research Institute, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Luna Djirackor
- Liverpool Ocular Oncology Research Group, University of Liverpool, Liverpool, United Kingdom
| | - Sarah E Coupland
- Liverpool Ocular Oncology Research Group, University of Liverpool, Liverpool, United Kingdom.,Liverpool Clinical Laboratories, Liverpool University Hospitals Foundation Trust, Liverpool, United Kingdom
| | - Helen Kalirai
- Liverpool Ocular Oncology Research Group, University of Liverpool, Liverpool, United Kingdom.,Liverpool Clinical Laboratories, Liverpool University Hospitals Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
36
|
Cho SY. Patient-derived xenografts as compatible models for precision oncology. Lab Anim Res 2020; 36:14. [PMID: 32461927 PMCID: PMC7238616 DOI: 10.1186/s42826-020-00045-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/06/2020] [Indexed: 01/02/2023] Open
Abstract
Cancer is a very heterogeneous disease, displaying heterogeneity between patients (inter-tumoral heterogeneity) and heterogeneity within a patient (intra-tumoral heterogeneity). Precision oncology is a diagnostic and therapeutic approach for cancers based on the stratification of patients using genomic and molecular profiling of tumors. To develop diagnostic and therapeutic tools for the application of precision oncology, appropriate preclinical mouse models that reflect tumor heterogeneity are required. Patient-derived xenograft (PDX) models are generated by the engraftment of patient tumors into immunodeficient mice that retain several aspects of the patient’s tumor characteristics, including inter-tumoral heterogeneity and intra-tumoral heterogeneity. Therefore, PDX models can be applied in various developmental steps of cancer diagnostics and therapeutics, such as biomarker development, companion diagnostics, drug efficacy testing, overcoming drug resistance, and co-clinical trials. This review summarizes the diverse aspects of PDX models, addressing the factors considered for PDX generation, application of PDX models for cancer research, and future directions of PDX models.
Collapse
Affiliation(s)
- Sung-Yup Cho
- 1Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080 South Korea.,2Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.,3Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, South Korea
| |
Collapse
|
37
|
Jin B, Zhang P, Zou H, Ye H, Wang Y, Zhang J, Yang H, Pan J. Verification of EZH2 as a druggable target in metastatic uveal melanoma. Mol Cancer 2020; 19:52. [PMID: 32127003 PMCID: PMC7055080 DOI: 10.1186/s12943-020-01173-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 02/24/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Hepatic metastasis develops in ~ 50% of uveal melanoma (UM) patients with no effective treatments. Although GNAQ/GNA11 mutations are believed to confer pathogenesis of UM, the underlying mechanism of liver metastasis remains poorly understood. Given that profound epigenetic evolution may occur in the long journey of circulating tumor cells (CTCs) to distant organs, we hypothesized that EZH2 endowed tumor cells with enhanced malignant features (e.g., stemness and motility) during hepatic metastasis in UM. We aimed to test this hypothesis and explore whether EZH2 was a therapeutic target for hepatic metastatic UM patients. METHODS Expression of EZH2 in UM was detected by qRT-PCR, Western blotting and immunohistochemistry staining. Proliferation, apoptosis, cancer stem-like cells (CSCs) properties, migration and invasion were evaluated under circumstances of treatment with either EZH2 shRNA or EZH2 inhibitor GSK126. Antitumor activity and frequency of CSCs were determined by xenografted and PDX models with NOD/SCID mice. Hepatic metastasis was evaluated with NOG mice. RESULTS We found that EZH2 overexpressed in UM promoted the growth of UM; EZH2 increased the percentage and self-renewal of CSCs by miR-29c-DVL2-β-catenin signaling; EZH2 facilitates migration and invasion of UM cells via RhoGDIγ-Rac1 axis. Targeting EZH2 either by genetics or small molecule inhibitor GSK126 decreased CSCs and motility and abrogated the liver metastasis of UM. CONCLUSIONS These findings validate EZH2 as a druggable target in metastatic UM patients, and may shed light on the understanding and interfering the complicated metastatic process.
Collapse
Affiliation(s)
- Bei Jin
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 South Xianlie Road, Guangzhou, 510060, People's Republic of China
| | - Ping Zhang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 South Xianlie Road, Guangzhou, 510060, People's Republic of China
| | - Hailin Zou
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 South Xianlie Road, Guangzhou, 510060, People's Republic of China
| | - Huijing Ye
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 South Xianlie Road, Guangzhou, 510060, People's Republic of China
| | - Yun Wang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 South Xianlie Road, Guangzhou, 510060, People's Republic of China
| | - Jing Zhang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 South Xianlie Road, Guangzhou, 510060, People's Republic of China
| | - Huasheng Yang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 South Xianlie Road, Guangzhou, 510060, People's Republic of China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 South Xianlie Road, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
38
|
Richards JR, Yoo JH, Shin D, Odelberg SJ. Mouse models of uveal melanoma: Strengths, weaknesses, and future directions. Pigment Cell Melanoma Res 2020; 33:264-278. [PMID: 31880399 PMCID: PMC7065156 DOI: 10.1111/pcmr.12853] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 12/21/2019] [Indexed: 12/14/2022]
Abstract
Uveal melanoma is the most common primary malignancy of the eye, and a number of discoveries in the last decade have led to a more thorough molecular characterization of this cancer. However, the prognosis remains dismal for patients with metastases, and there is an urgent need to identify treatments that are effective for this stage of disease. Animal models are important tools for preclinical studies of uveal melanoma. A variety of models exist, and they have specific advantages, disadvantages, and applications. In this review article, these differences are explored in detail, and ideas for new models that might overcome current challenges are proposed.
Collapse
Affiliation(s)
- Jackson R. Richards
- Department of Oncological SciencesUniversity of UtahSalt Lake CityUTUSA
- Program in Molecular MedicineUniversity of UtahSalt Lake CityUTUSA
| | - Jae Hyuk Yoo
- Program in Molecular MedicineUniversity of UtahSalt Lake CityUTUSA
| | - Donghan Shin
- Program in Molecular MedicineUniversity of UtahSalt Lake CityUTUSA
| | - Shannon J. Odelberg
- Program in Molecular MedicineUniversity of UtahSalt Lake CityUTUSA
- Department of Internal MedicineDivision of Cardiovascular MedicineUniversity of UtahSalt Lake CityUTUSA
- Department of Neurobiology and AnatomyUniversity of UtahSalt Lake CityUTUSA
| |
Collapse
|
39
|
Okano M, Oshi M, Butash A, Okano I, Saito K, Kawaguchi T, Nagahashi M, Kono K, Ohtake T, Takabe K. Orthotopic Implantation Achieves Better Engraftment and Faster Growth Than Subcutaneous Implantation in Breast Cancer Patient-Derived Xenografts. J Mammary Gland Biol Neoplasia 2020; 25:27-36. [PMID: 32109311 PMCID: PMC7141774 DOI: 10.1007/s10911-020-09442-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 01/22/2020] [Indexed: 01/18/2023] Open
Abstract
Patient-Derived Xenograft (PDX) is now accepted as a murine model that better mimics human cancer when compared to a conventional cancer cell-line inoculation model. Some claim the advantage of orthotopic site implantation of patient tumor (OS) over ectopic implantation into the subcutaneous space (SQ); however, there has been no study that describes a head-to-head comparison of oncological differences between these two models to date. We hypothesize that OS tumors re-transplant and grow better than SQ tumors and are therefore a better model to evaluate tumor aggressiveness. Breast cancer PDXs were generated using the tumors derived from 11 patients into NOD scid gamma (NSG) mice. We used six ER(+)HER2(-) tumors and five triple negative (TN) tumors for a total of 11 tumors. Five PDX lines grew for an overall engraftment rate of 45%. We present our OS implantation method in detail. The re-transplantation rate of TN tumors in each transplant site was significantly higher in OS when compared to SQ tumors (70.1% vs. 32.1%, p < 0.01). OS tumors grow significantly faster than SQ tumors. Similarly, OS tumors demonstrated significantly more mitotic figures and Ki-67 positive cells than SQ tumors. The tumor re-transplantation rate significantly increased by the second and third generations with the OS method. The time from implantation to development of a palpable tumor dramatically decreased after the first passage. PDX of ER(+) tumors demonstrated significantly lower engraftment rates and slower tumor growth than TN tumors, which remarkably improved by the first passage. Orthotopically implanted PDX tumors showed better re-transplantation rates, greater tumor size, and more significant growth compared to the subcutaneously implanted model.
Collapse
Affiliation(s)
- Maiko Okano
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Masanori Oshi
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
| | - Ali Butash
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
| | - Ichiro Okano
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
| | - Katsuharu Saito
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tsutomu Kawaguchi
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
| | - Masayuki Nagahashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Toru Ohtake
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kazuaki Takabe
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA.
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan.
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY, USA.
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan.
- Department of Surgery, Yokohama City University, Yokohama, Japan.
| |
Collapse
|
40
|
Kageyama K, Ozaki S, Sato T. Generation of a Liver Orthotopic Human Uveal Melanoma Xenograft Platform in Immunodeficient Mice. J Vis Exp 2019. [PMID: 31762467 DOI: 10.3791/59941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
In recent decades, subcutaneously implanted patient-derived xenograft tumors or cultured human cell lines have been increasingly recognized as more representative models to study human cancers in immunodeficient mice than traditional established human cell lines in vitro. Recently, orthotopically implanted patient-derived tumor xenograft (PDX) models in mice have been developed to better replicate features of patient tumors. A liver orthotopic xenograft mouse model is expected to be a useful cancer research platform, providing insights into tumor biology and drug therapy. However, liver orthotopic tumor implantation is generally complicated. Here we describe our protocols for the orthotopic implantation of patient-derived liver-metastatic uveal melanoma tumors. We cultured human liver metastatic uveal melanoma cell lines into immunodeficient mice. The protocols can result in consistently high technical success rates using either a surgical orthotopic implantation technique with chunks of patient-derived uveal melanoma tumor or a needle injection technique with cultured human cell line. We also describe protocols for CT scanning to detect interior liver tumors and for re-implantation techniques using cryopreserved tumors to achieve re-engraftment. Together, these protocols provide a better platform for liver orthotopic tumor mouse models of liver metastatic uveal melanoma in translational research.
Collapse
Affiliation(s)
- Ken Kageyama
- Department of Diagnostic and Interventional Radiology, Osaka City University Graduate School of Medicine; Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University
| | - Shinji Ozaki
- Department of Surgery, National Hospital Organization, Kure Medical Cancer Center; Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University
| | - Takami Sato
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University;
| |
Collapse
|
41
|
Zhang J, Liu S, Ye Q, Pan J. Transcriptional inhibition by CDK7/9 inhibitor SNS-032 abrogates oncogene addiction and reduces liver metastasis in uveal melanoma. Mol Cancer 2019; 18:140. [PMID: 31526394 PMCID: PMC6745806 DOI: 10.1186/s12943-019-1070-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022] Open
Abstract
Background Life of patients with uveal melanoma (UM) is largely threatened by liver metastasis. Little is known about the drivers of liver organotropic metastasis in UM. The elevated activity of transcription of oncogenes is presumably to drive aspects of tumors. We hypothesized that inhibition of transcription by cyclin-dependent kinase 7/9 (CDK7/9) inhibitor SNS-032 diminished liver metastasis by abrogating the putative oncogenes in charge of colonization, stemness, cell motility of UM cells in host liver microenvironment. Methods The effects of SNS-032 on the expression of the relevant oncogenes were examined by qRT-PCR and Western blotting analysis. Proliferative activity, frequency of CSCs and liver metastasis were evaluated by using NOD-SCID mouse xenograft model and NOG mouse model, respectively. Results The results showed that CDK7/9 were highly expressed in UM cells, and SNS-032 significantly suppressed the cellular proliferation, induced apoptosis, and inhibited the outgrowth of xenografted UM cells and PDX tumors in NOD-SCID mice, repressed the cancer stem-like cell (CSC) properties through transcriptional inhibition of stemness-related protein Krüppel-like factor 4 (KLF4), inhibited the invasive phonotypes of UM cells through matrix metalloproteinase 9 (MMP9). Mechanistically, SNS-032 repressed the c-Myc-dependent transcription of RhoA gene, and thereby lowered the RhoA GTPase activity and actin polymerization, and subsequently inhibited cell motility and liver metastasis. Conclusions In conclusion, we validate a set of transcription factors which confer metastatic traits (e.g., KLF4 for CSCs, c-Myc for cell motility) in UM cells. Our results identify SNS-032 as a promising therapeutic agent, and warrant a clinical trial in patients with metastatic UM.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 South Xianlie Road, Guangzhou, 510060, People's Republic of China
| | - Shenglan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 South Xianlie Road, Guangzhou, 510060, People's Republic of China
| | - Qianyun Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 South Xianlie Road, Guangzhou, 510060, People's Republic of China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 South Xianlie Road, Guangzhou, 510060, People's Republic of China
| |
Collapse
|
42
|
Bray LJ, Hutmacher DW, Bock N. Addressing Patient Specificity in the Engineering of Tumor Models. Front Bioeng Biotechnol 2019; 7:217. [PMID: 31572718 PMCID: PMC6751285 DOI: 10.3389/fbioe.2019.00217] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer treatment is challenged by the heterogeneous nature of cancer, where prognosis depends on tumor type and disease stage, as well as previous treatments. Optimal patient stratification is critical for the development and validation of effective treatments, yet pre-clinical model systems are lacking in the delivery of effective individualized platforms that reflect distinct patient-specific clinical situations. Advances in cancer cell biology, biofabrication, and microengineering technologies have led to the development of more complex in vitro three-dimensional (3D) models to act as drug testing platforms and to elucidate novel cancer mechanisms. Mostly, these strategies have enabled researchers to account for the tumor microenvironment context including tumor-stroma interactions, a key factor of heterogeneity that affects both progression and therapeutic resistance. This is aided by state-of-the-art biomaterials and tissue engineering technologies, coupled with reproducible and high-throughput platforms that enable modeling of relevant physical and chemical factors. Yet, the translation of these models and technologies has been impaired by neglecting to incorporate patient-derived cells or tissues, and largely focusing on immortalized cell lines instead, contributing to drug failure rates. While this is a necessary step to establish and validate new models, a paradigm shift is needed to enable the systematic inclusion of patient-derived materials in the design and use of such models. In this review, we first present an overview of the components responsible for heterogeneity in different tumor microenvironments. Next, we introduce the state-of-the-art of current in vitro 3D cancer models employing patient-derived materials in traditional scaffold-free approaches, followed by novel bioengineered scaffold-based approaches, and further supported by dynamic systems such as bioreactors, microfluidics, and tumor-on-a-chip devices. We critically discuss the challenges and clinical prospects of models that have succeeded in providing clinical relevance and impact, and present emerging concepts of novel cancer model systems that are addressing patient specificity, the next frontier to be tackled by the field.
Collapse
Affiliation(s)
- Laura J. Bray
- School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
- Translational Research Institute, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Dietmar W. Hutmacher
- School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
- Translational Research Institute, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane, QLD, Australia
- Australian Research Council (ARC) Industrial Transformation Training Centre in Additive Biomanufacturing, Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
| | - Nathalie Bock
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
- Translational Research Institute, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane, QLD, Australia
| |
Collapse
|
43
|
PARP Inhibition Increases the Response to Chemotherapy in Uveal Melanoma. Cancers (Basel) 2019; 11:cancers11060751. [PMID: 31146482 PMCID: PMC6628115 DOI: 10.3390/cancers11060751] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/16/2019] [Accepted: 05/27/2019] [Indexed: 12/18/2022] Open
Abstract
Uveal melanoma (UM) remains without effective therapy at the metastatic stage, which is associated with BAP-1 (BRCA1 associated protein) mutations. However, no data on DNA repair capacities in UM are available. Here, we use UM patient-derived xenografts (PDXs) to study the therapeutic activity of the PARP inhibitor olaparib, alone or in combination. First, we show that the expression and the activity of PARP proteins is similar between the PDXs and the corresponding patient’s tumors. In vivo experiments in the PDX models showed that olaparib was not efficient alone, but significantly increased the efficacy of dacarbazine. Finally, using reverse phase protein arrays and immunohistochemistry, we identified proteins involved in DNA repair and apoptosis as potential biomarkers predicting response to the combination of olaparib and dacarbazine. We also observed a high increase of phosphorylated YAP and TAZ proteins after dacarbazine + olaparib treatment. Our results suggest that PARP inhibition in combination with the alkylating agent dacarbazine could be of clinical interest for UM treatment. We also observe an interesting effect of dacarbazine on the Hippo pathway, confirming the importance of this pathway in UM.
Collapse
|
44
|
Establishment and characterization of melanoma patient-derived xenograft models for preclinical evaluation of novel therapeutics. Melanoma Res 2019; 28:527-535. [PMID: 30086074 DOI: 10.1097/cmr.0000000000000494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Patient-derived xenograft (PDX) models mostly retain the histological and genetic features of their donor tumors, which have been used for investigating various types of cancer. However, PDX models for melanoma, especially acral melanoma, are reported occasionally. We aimed to establish a large panel of melanoma PDX models representing the predominant Asian melanomas. Ninety-three fresh melanoma samples were implanted subcutaneously into nonobese diabetic/severe combined immunodeficiency mice. The histological and genetic characteristics were analyzed in both patient tumors and PDX models using immunohistochemistry, PCR amplification, and Sanger sequencing. Furthermore, the sensitivities of PDX models harboring distinct mutation profiles to binimetinib (a MEK inhibitor), vemubrafenib (a BRAF inhibitor), and imatinib (a KIT inhibitor) were also evaluated. Twenty-five PDX models were established successfully [25/93 (26.9%)] and passaged to maintain tumors in vivo. Clinical stage and origin of tumor sample were correlated with successful establishment rates (P=0.008 and <0.001, respectively). The histological (expression of NRAS, P16, and RB) and genetic (mutation status of NRAS, BRAF, and KIT) characteristics were stably maintained from patient tumors to PDX models. Targeted drugs could inhibit the tumor growth of PDX models harboring the corresponding target gene mutations. These PDX models constitute a pharmacological platform, enabling personalized development of therapeutic strategies for Asian melanomas.
Collapse
|
45
|
Cornillie J, Wozniak A, Li H, Wang Y, Boeckx B, Gebreyohannes YK, Wellens J, Vanleeuw U, Hompes D, Stas M, Sinnaeve F, Wafa H, Lambrechts D, Debiec-Rychter M, Sciot R, Schöffski P. Establishment and Characterization of Histologically and Molecularly Stable Soft-tissue Sarcoma Xenograft Models for Biological Studies and Preclinical Drug Testing. Mol Cancer Ther 2019; 18:1168-1178. [PMID: 30962320 DOI: 10.1158/1535-7163.mct-18-1045] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/13/2019] [Accepted: 04/01/2019] [Indexed: 11/16/2022]
Abstract
Soft-tissue sarcomas (STS) represent a heterogeneous group of rare, malignant tumors of mesenchymal origin. Reliable in vivo sarcoma research models are scarce. We aimed to establish and characterize histologically and molecularly stable patient-derived xenograft (PDX) models from a broad variety of STS subtypes. A total of 188 fresh tumor samples from consenting patients with localized or advanced STS were transplanted subcutaneously in NMRI-nu/nu-immunodeficient mice. Once tumor growth was observed, the material was passaged to a next generation of mice. A patient-derived tumor sample was considered "successfully engrafted" whenever the sample was transplanted to passage 1. A PDX model was considered "established" when observing stable morphologic and molecular features for at least two passages. With every passage, histologic and molecular analyses were performed. Specific genomic alterations and copy-number profile were assessed by FISH and low coverage whole-genome sequencing. The tumor engraftment rate was 32% (61/188) and 188 patient samples generated a total of 32 PDX models, including seven models of myxofibrosarcoma, five dedifferentiated liposarcoma, five leiomyosarcoma, three undifferentiated pleomorphic sarcoma, two malignant peripheral nerve sheet tumor models, and single models of synovial sarcoma and some other (ultra)rare subtypes. Seventeen additional models are in early stages of engraftment (passage 1-2). Histopathologic and molecular features were compared with the original donor tumor and were stable throughout passaging. The platform is used for studies on sarcoma biology and suited for in vivo preclinical drug testing as illustrated by a number of completed and ongoing laboratory studies.
Collapse
Affiliation(s)
- Jasmien Cornillie
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Agnieszka Wozniak
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Haifu Li
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Yannick Wang
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Bram Boeckx
- Laboratory for Translational Genetics, KU Leuven, Leuven, Belgium.,VIB Center for Cancer Biology, Leuven, Belgium
| | - Yemarshet K Gebreyohannes
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Jasmien Wellens
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium.
| | - Ulla Vanleeuw
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Daphne Hompes
- Department of Surgical Oncology, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Marguerite Stas
- Department of Surgical Oncology, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Friedl Sinnaeve
- Department of Orthopedic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Hazem Wafa
- Department of Orthopedic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, KU Leuven, Leuven, Belgium.,VIB Center for Cancer Biology, Leuven, Belgium
| | - Maria Debiec-Rychter
- Department of Human Genetics, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Raf Sciot
- Department of Pathology, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Patrick Schöffski
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| |
Collapse
|
46
|
Malcolm JE, Stearns TM, Airhart SD, Graber JH, Bult CJ. Factors that influence response classifications in chemotherapy treated patient-derived xenografts (PDX). PeerJ 2019; 7:e6586. [PMID: 30944774 PMCID: PMC6441558 DOI: 10.7717/peerj.6586] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 02/08/2019] [Indexed: 01/06/2023] Open
Abstract
In this study, we investigated the impact of initial tumor volume, rate of tumor growth, cohort size, study duration, and data analysis method on chemotherapy treatment response classifications in patient-derived xenografts (PDXs). The analyses were conducted on cisplatin treatment response data for 70 PDX models representing ten cancer types with up to 28-day study duration and cohort sizes of 3-10 tumor-bearing mice. The results demonstrated that a 21-day dosing study using a cohort size of eight was necessary to reliably detect responsive models (i.e., tumor volume ratio of treated animals to control between 0.1 and 0.42)-independent of analysis method. A cohort of three tumor-bearing animals led to a reliable classification of models that were both highly responsive and highly nonresponsive to cisplatin (i.e., tumor volume ratio of treated animals to control animals less than 0.10). In our set of PDXs, we found that tumor growth rate in the control group impacted treatment response classification more than initial tumor volume. We repeated the study design factors using docetaxel treated PDXs with consistent results. Our results highlight the importance of defining endpoints for PDX dosing studies when deciding the size of cohorts to use in dosing studies and illustrate that response classifications for a study do not differ significantly across the commonly used analysis methods that are based on tumor volume changes in treatment versus control groups.
Collapse
Affiliation(s)
- Joan E. Malcolm
- The Jackson Laboratory, Bar Harbor, ME, United States of America
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States of America
| | | | - Susan D. Airhart
- The Jackson Laboratory, Bar Harbor, ME, United States of America
| | - Joel H. Graber
- The Jackson Laboratory, Bar Harbor, ME, United States of America
- The MDI Biological Laboratory, Bar Harbor, ME, United States of America
| | - Carol J. Bult
- The Jackson Laboratory, Bar Harbor, ME, United States of America
| |
Collapse
|
47
|
Yang CY, Lin CK, Hsieh CC, Tsao CH, Lin CS, Peng B, Chen YT, Ting CC, Chang WC, Lin GJ, Sytwu HK, Chen YW. Anti-oral cancer effects of triptolide by downregulation of DcR3 in vitro, in vivo, and in preclinical patient-derived tumor xenograft model. Head Neck 2018; 41:1260-1269. [PMID: 30537218 PMCID: PMC6590365 DOI: 10.1002/hed.25554] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 10/08/2018] [Accepted: 10/31/2018] [Indexed: 12/15/2022] Open
Abstract
Background Aberrant expression of decoy receptor 3 (DcR3) is considered to be a diagnostic and therapeutic target for human cancers. The aim of this study was to assess DcR3 as a target of the anticancer effects of triptolide (TPL) in preclinical patient‐derived tumor xenograft (PDTX) models of oral squamous cell carcinoma (OSCC). Methods The expression of DcR3 was evaluated through immunohistochemistry, and correlations were examined using clinical variables. The effects of TPL on the expression of DcR3 and cell proliferation were investigated in OSCC cell lines and in PDTX models. Results DcR3 overexpression was associated with overall survival and tumor size. TPL significantly decreased tumor growth. Moreover, TPL inhibited the expression of metastasis‐associated protein 1 (MTA1), a transcription factor for DcR3 in vivo, in vitro, and in PDTX models. Conclusion TPL appeared to exert anticancer effects by repressing DcR3 and MTA1 in vitro, in vivo, and in PDTX models.
Collapse
Affiliation(s)
- Cheng-Yu Yang
- School of Dentistry, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Kung Lin
- Division of Anatomic Pathology, Taipei Tzu Chi Hospital, Taipei, Taiwan
| | - Cheng-Chih Hsieh
- Department of Pharmacy Practice, Tri-Service General Hospital, Taipei, Taiwan
| | - Chang-Huei Tsao
- Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan.,Department of Medical Research, Tri-Service General Hospital, Taipei, Taiwan
| | - Chun-Shu Lin
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Centre, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Bo Peng
- School of Dentistry, National Defense Medical Center, Taipei, Taiwan
| | - Yen-Tzu Chen
- School of Dentistry, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Chieh Ting
- School of Dentistry, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Chin Chang
- School of Dentistry, National Defense Medical Center, Taipei, Taiwan.,Department of Oral and Maxillofacial Surgery, Tri-Service General Hospital, Taipei, Taiwan
| | - Gu-Jiun Lin
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Huey-Kang Sytwu
- Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Yuan-Wu Chen
- School of Dentistry, National Defense Medical Center, Taipei, Taiwan.,Department of Oral and Maxillofacial Surgery, Tri-Service General Hospital, Taipei, Taiwan
| |
Collapse
|
48
|
Abstract
Immunotherapy is one of the most exciting recent breakthroughs in the field of cancer treatment. Many different approaches are being developed and a number have already gained regulatory approval or are under investigation in clinical trials. However, learning from the past, preclinical animal models often insufficiently reflect the physiological situation in humans, which subsequently causes treatment failures in clinical trials. Due to species-specific differences in most parts of the immune system, the transfer of knowledge from preclinical studies to clinical trials is eminently challenging. Human tumor cell line-based or patient-derived xenografts in immunocompromised mice have been successfully applied in the preclinical testing of cytotoxic or molecularly targeted agents, but naturally these systems lack the human immune system counterpart. The co-transplantation of human peripheral blood mononuclear cells or hematopoietic stem cells is employed to overcome this limitation. This review summarizes some important aspects of the different available tumor xenograft mouse models, their history, and their implementation in drug development and personalized therapy. Moreover, recent progress, opportunities and limitations of different humanized mouse models will be discussed.
Collapse
|
49
|
Zhu H, Wang C, Wang J, Chen D, Deng J, Deng J, Fan J, Badakhshi H, Huang X, Zhang L, Cai J, Guo S, Qian W, Nie Y, Li Q, Zhao K. A subset of esophageal squamous cell carcinoma patient-derived xenografts respond to cetuximab, which is predicted by high EGFR expression and amplification. J Thorac Dis 2018; 10:5328-5338. [PMID: 30416780 DOI: 10.21037/jtd.2018.09.18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Epidermal growth factor receptor (EGFR) is reportedly overexpressed in most esophageal tumors, but most targeted therapies showed no efficacy in non-selected patients. This study aims at investigating the adaptive cetuximab subset in a cohort of esophageal squamous cell carcinoma (ESCC) patient-derived xenografts (PDXs). Methods A large panel of ESCC PDXs has been established. The copy number, mRNA expression and immunohistochemistry (IHC) of key EGFR pathways have been examined along with cetuximab response. A preclinical trial on a randomly selected cohort of 16 ESCC PDXs was conducted, and the genomic annotations of these models were compared against the efficacy readout of the mouse trial. Results The trial identified that 7 of 16 (43.8%) responded to cetuximab (ΔT/ΔC <0 as responders). The gene amplification and expression analysis indicated that EGFR copy number ≥5 (P=0.035), high EGFR mRNA expression (P=0.001) and IHC score of 2-3 (P=0.034) are associated with tumor growth inhibition by cetuximab, suggesting EGFR may function as a single predictive biomarker for cetuximab response in ESCC. Conclusions Overall, our results suggest that an ESCC subtype with EGFR amplification and overexpression benefits from cetuximab treatment, which warrants further clinical confirmation.
Collapse
Affiliation(s)
- Hanting Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chunyu Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | | | - Dawei Chen
- Crown Bioscience, Inc., San Diego, CA, USA
| | - Jiaying Deng
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | | | - Jianhong Fan
- Department of Gynaecology, Renhe Hospital, Shanghai 200431, China
| | - Harun Badakhshi
- Department of Radiation Oncology, Charité School of Medicine and Centre for Cancer Medicine, Berlin, Germany
| | | | | | - Jie Cai
- Crown Bioscience, Inc., San Diego, CA, USA
| | - Sheng Guo
- Crown Bioscience, Inc., San Diego, CA, USA
| | - Wubin Qian
- Crown Bioscience, Inc., San Diego, CA, USA
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digest Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Qixiang Li
- Crown Bioscience, Inc., San Diego, CA, USA.,State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Kuaile Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
50
|
Slater K, Hoo PS, Buckley AM, Piulats JM, Villanueva A, Portela A, Kennedy BN. Evaluation of oncogenic cysteinyl leukotriene receptor 2 as a therapeutic target for uveal melanoma. Cancer Metastasis Rev 2018; 37:335-345. [DOI: 10.1007/s10555-018-9751-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|