1
|
Ferrier ST, Li M, Burnier JV. Azacytidine treatment affects the methylation pattern of genomic and cell-free DNA in uveal melanoma cell lines. BMC Cancer 2024; 24:1299. [PMID: 39434038 PMCID: PMC11495039 DOI: 10.1186/s12885-024-13037-4] [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: 05/03/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Uveal melanoma (UM) is the most common primary intraocular tumour in adults, and approximately 50% of patients will develop metastasis. Epigenetic changes are a major factor in cancer progression. We aimed to determine whether methylation profiles could be altered using a DNA methyltransferase (DNMT) inhibitor in UM cell lines. METHODS Four primary and metastatic UM cell lines were treated with azacytidine and analysed for cell proliferation, colony formation, and BAP1 protein expression. Genomic and cell-free (cf)DNA methylation were compared. RESULTS In all cell lines, azacytidine treatment resulted in dose-dependent effects on proliferation, colony formation, and radiosensitivity. Methylation profiling revealed differences in methylation between cell lines according to BAP1 expression. Matched primary and metastatic cell lines showed very similar patterns. Alterations were seen in pathways known to be important in UM progression, such as PI3K/Akt and MAPK signaling, and in pathways involved in cancer progression, such as regulation of stemlike potential, cell motility, and invasion. These changes were maintained in genomic and cell-free DNA. CONCLUSIONS This data suggests that DNMT inhibitors cause changes in UM cells that are maintained in cfDNA. The results suggest that targeting methylation in UM treatment and monitoring response to treatment using cfDNA methylation could be a valuable tool.
Collapse
Affiliation(s)
- Sarah Tadhg Ferrier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Québec, Canada
- Department of Pathology, McGill University, Montreal, Québec, Canada
| | - Mingyang Li
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Québec, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Québec, Canada.
- Department of Pathology, McGill University, Montreal, Québec, Canada.
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
2
|
Zhu X, Wang L, Wang K, Yao Y, Zhou F. Erdafitinib promotes ferroptosis in human uveal melanoma by inducing ferritinophagy and lysosome biogenesis via modulating the FGFR1/mTORC1/TFEB signaling axis. Free Radic Biol Med 2024; 222:552-568. [PMID: 38971541 DOI: 10.1016/j.freeradbiomed.2024.07.002] [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: 05/14/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Uveal melanoma (UM) is a rare yet lethal primary intraocular malignancy affecting adults. Analysis of data from The Cancer Genome Atlas (TCGA) database revealed that FGFR1 expression was increased in UM tumor tissues and was linked to aggressive behavior and a poor prognosis. This study assessed the anti-tumor effects of Erdafitinib, a selective pan-FGFR inhibitor, in both in vitro and in vivo UM models. Erdafitinib exhibited a robust anti-cancer activity in UM through inducing ferroptosis in the FGFR1-dependent manner. Transcriptomic data revealed that Erdafitinib mediated its anti-cancer effects via modulating the ferritinophagy/lysosome biogenesis. Subsequent research revealed that Erdafitinib exerted its effects by reducing the expression of FGFR1 and inhibiting the activity of mTORC1 in UM cells. Concurrently, it enhanced the dephosphorylation, nuclear translocation, and transcriptional activity of TFEB. The aggregation of TFEB in nucleus triggered FTH1-dependent ferritinophagy, leading to lysosomal activation and iron overload. Conversely, the overexpression of FGFR1 served to mitigate the effects of Erdafitinib on ferritinophagy, lysosome biogenesis, and the activation of the mTORC1/TFEB signaling pathway. In vivo experiments have convincingly shown that Erdafitinib markedly curtails tumor growth in an UM xenograft mouse model, an effect that is closely correlated with a decrease in FGFR1 expression levels. The present study is the first to demonstrate that Erdafitinib powerfully induces ferroptosis in UM by orchestrating the ferritinophagy and lysosome biogenesis via modulating the FGFR1/mTORC1/TFEB signaling. Consequently, Erdafitinib emerges as a strong candidate for clinical trial investigation, and FGFR1 emerges as a novel and promising therapeutic target in the treatment of UM.
Collapse
Affiliation(s)
- Xue Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China
| | - Ling Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China
| | - Ke Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China.
| | - Ying Yao
- Department of Pharmacy, Wuxi Maternity and Child Health Care Hospital, Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, 214002, Jiangsu Province, China.
| | - Fanfan Zhou
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
3
|
Păsărică MA, Curcă PF, Dragosloveanu CDM, Grigorescu AC, Nisipașu CI. Pathological and Molecular Diagnosis of Uveal Melanoma. Diagnostics (Basel) 2024; 14:958. [PMID: 38732371 PMCID: PMC11083017 DOI: 10.3390/diagnostics14090958] [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: 02/29/2024] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
(1) Background: Uveal melanoma (UM) is a common malignant intraocular tumor that presents with significant genetic differences to cutaneous melanoma and has a high genetic burden in terms of prognosis. (2) Methods: A systematic literature search of several repositories on uveal melanoma diagnosis, prognosis, molecular analysis, and treatment was conducted. (3) Results: Recent genetic understanding of oncogene-initiation mutations in GNAQ, GNA11, PLCB4, and CYSLTR2 and secondary progression drivers of BAP1 inactivation and SF3B1 and EIF1AX mutations offers an appealing explanation to the high prognostic impact of adding genetic profiling to clinical UM classification. Genetic information could help better explain peculiarities in uveal melanoma, such as the low long-term survival despite effective primary tumor treatment, the overwhelming propensity to metastasize to the liver, and possibly therapeutic behaviors. (4) Conclusions: Understanding of uveal melanoma has improved step-by-step from histopathology to clinical classification to more recent genetic understanding of oncogenic initiation and progression.
Collapse
Affiliation(s)
- Mihai Adrian Păsărică
- Clinical Department of Ophthalmology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.A.P.); (C.D.M.D.)
- Department of Ophthalmology, Clinical Hospital for Ophthalmological Emergencies, 010464 Bucharest, Romania
| | - Paul Filip Curcă
- Clinical Department of Ophthalmology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.A.P.); (C.D.M.D.)
- Department of Ophthalmology, Clinical Hospital for Ophthalmological Emergencies, 010464 Bucharest, Romania
| | - Christiana Diana Maria Dragosloveanu
- Clinical Department of Ophthalmology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.A.P.); (C.D.M.D.)
- Department of Ophthalmology, Clinical Hospital for Ophthalmological Emergencies, 010464 Bucharest, Romania
| | | | - Cosmin Ionuț Nisipașu
- Department of Dental Medicine I, Implant-Prosthetic Therapy, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| |
Collapse
|
4
|
Bi HE, Zhang J, Yao Y, Wang S, Yao J, Shao Z, Jiang Q. Expression and functional significance of phosphoenolpyruvate carboxykinase 1 in uveal melanoma. Cell Death Discov 2024; 10:196. [PMID: 38670942 PMCID: PMC11053060 DOI: 10.1038/s41420-024-01963-y] [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: 01/28/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Uveal melanoma (UVM), an uncommon yet potentially life-threatening ocular cancer, arises from melanocytes in the uveal tract of the eye. The exploration of novel oncotargets for UVM is of paramount importance. In this study, we show that PCK1 (phosphoenolpyruvate carboxykinase 1) expression is upregulated in various UVM tissues as well as in primary UVM cells and immortalized lines. Furthermore, bioinformatics studies reveal that PCK1 overexpression in UVM correlates with advanced disease stages and poor patient survival. Genetic silencing (utilizing viral shRNA) or knockout (via CRISPR/Cas9) of PCK1 significantly curtailed cell viability, proliferation, cell cycle progression, and motility, while provoking apoptosis in primary and immortalized UVM cells. Conversely, ectopic overexpression of PCK1, achieved through a viral construct, bolstered UVM cell proliferation and migration. Gαi3 expression and Akt phosphorylation were reduced following PCK1 silencing or knockout, but increased after PCK1 overexpression in UVM cells. Restoring Akt phosphorylation through a constitutively active mutant Akt1 (S473D) ameliorated the growth inhibition, migration suppression, and apoptosis induced by PCK1 silencing in UVM cells. Additionally, ectopic expression of Gαi3 restored Akt activation and counteracted the anti-UVM cell effects by PCK1 silencing. In vivo, the growth of subcutaneous xenografts of primary human UVM cells was significantly inhibited following intratumoral injection of adeno-associated virus (aav) expressing PCK1 shRNA. PCK1 depletion, Gαi3 downregulation, Akt inhibition, proliferation arrest, and apoptosis were detected in PCK1-silenced UVM xenografts. Collectively, our findings demonstrate that PCK1 promotes UVM cell growth possibly by modulating the Gαi3-Akt signaling pathway.
Collapse
Affiliation(s)
- Hui-E Bi
- The Affiliated Eye Hospital, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Department of Ophthalmology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jie Zhang
- Obstetrics and Gynecology Department, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Yujia Yao
- The Affiliated Eye Hospital, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Suyu Wang
- The Affiliated Eye Hospital, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Jin Yao
- The Affiliated Eye Hospital, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
| | - Zhijiang Shao
- Department of Ophthalmology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.
| | - Qin Jiang
- The Affiliated Eye Hospital, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
5
|
Gunenc D, Ozluk AA, Yıldırım UM, Ascierto PA, Karaca B. Successful application of chemosaturation with percutaneous hepatic perfusion in metastatic uveal melanoma patient progressing after systemic treatment options: a case report. Front Oncol 2024; 14:1355971. [PMID: 38660135 PMCID: PMC11040682 DOI: 10.3389/fonc.2024.1355971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Uveal melanoma (UM) is a rare subtype of melanoma, accounting for less than 5% of all melanoma cases. Metastatic UM differs notably from cutaneous melanoma, exhibiting variations in etiology, prognosis, driver mutations, metastatic patterns, and poor responses to immune checkpoint inhibitors (ICI). Beyond local treatment options, such as resection, radiation therapy, and enucleation, and systemic treatments, such as ICIs, the approval of tebentafusp, a bispecific gp100 peptide-HLA-directed CD3 T-cell engager, marks a breakthrough in treating HLA-A*02:01 metastatic UM. Despite the advancements in treatment options, the long-term survival rates remain inadequate. We report a patient with metastatic UM who previously received ICI and progressed on tebentafusp treatment but subsequently exhibited a remarkable response to local treatment targeting liver metastasis. Such observations highlight the significance of exploring sequential therapeutic strategies for advanced UM, offering potential avenues to enhance treatment efficacy and patient prognosis.
Collapse
Affiliation(s)
- Damla Gunenc
- Department of Medical Oncology, Ege University Faculty of Medicine, Izmir, Türkiye
| | - Ahmet Anil Ozluk
- Department of Medical Oncology, Ege University Faculty of Medicine, Izmir, Türkiye
| | - Utku Mahir Yıldırım
- Department of Interventional Radiology, Izmir University of Economics, Medicalpoint Hospital, Izmir, Türkiye
| | - Paolo A. Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale dei Tumori IRCCS “Fondazione G. Pascale”, Naples, Italy
| | - Burcak Karaca
- Department of Medical Oncology, Ege University Faculty of Medicine, Izmir, Türkiye
| |
Collapse
|
6
|
Li Y, Zhu T, Yang J, Zhang Q, Xu S, Ge S, Jia R, Zhang J, Fan X. EHMT2 promotes tumorigenesis in GNAQ/11-mutant uveal melanoma via ARHGAP29-mediated RhoA pathway. Acta Pharm Sin B 2024; 14:1187-1203. [PMID: 38486999 PMCID: PMC10935147 DOI: 10.1016/j.apsb.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/30/2023] [Accepted: 12/02/2023] [Indexed: 03/17/2024] Open
Abstract
Constitutive activation of GNAQ/11 is the initiative oncogenic event in uveal melanoma (UM). Direct targeting GNAQ/11 has yet to be proven feasible as they are vital for a plethora of cellular functions. In search of genetic vulnerability for UM, we found that inhibition of euchromatic histone lysine methyltransferase 2 (EHMT2) expression or activity significantly reduced the proliferation and migration capacity of cancer cells. Notably, elevated expression of EHMT2 had been validated in UM samples. Furthermore, Kaplan-Meier survival analysis indicated high EHMT2 protein level was related to poor recurrence-free survival and a more advanced T stage. Chromatin immunoprecipitation sequencing analysis and the following mechanistic investigation showed that ARHGAP29 was a downstream target of EHMT2. Its transcription was suppressed by EHMT2 in a methyltransferase-dependent pattern in GNAQ/11-mutant UM cells, leading to elevated RhoA activity. Rescuing constitutively active RhoA in UM cells lacking EHMT2 restored oncogenic phenotypes. Simultaneously blocking EHMT2 and GNAQ/11 signaling in vitro and in vivo showed a synergistic effect on UM growth, suggesting the driver role of these two key molecules. In summary, our study shows evidence for an epigenetic program of EHMT2 regulation that influences UM progression and indicates inhibiting EHMT2 and MEK/ERK simultaneously as a therapeutic strategy in GNAQ/11-mutant UM.
Collapse
Affiliation(s)
- Yongyun Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Tianyu Zhu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Jie Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Qianqian Zhang
- National Research Center for Translational Medicine (Shanghai), State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shiqiong Xu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| | - Jianming Zhang
- Institute of Translational Medicine, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200001, China
| |
Collapse
|
7
|
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
|
8
|
Miley DR, Andrews-Pfannkoch CM, Pulido JS, Erickson SA, Vile RG, Fautsch MP, Marmorstein AD, Dalvin LA. Direct early growth response-1 knockdown decreases melanoma viability independent of mitogen-activated extracellular signal-related kinase inhibition. Melanoma Res 2023; 33:482-491. [PMID: 37650708 PMCID: PMC10615778 DOI: 10.1097/cmr.0000000000000921] [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: 09/01/2023]
Abstract
To investigate downstream molecular changes caused by mitogen-activated protein kinase (MEK) inhibitor treatment and further explore the impact of direct knockdown of early growth response-1 ( EGR1 ) in melanoma cell culture. RNA-sequencing (RNA-Seq) was performed to determine gene expression changes with MEK inhibitor treatment. Treatment with MEK inhibitor (trametinib) was then assessed in two cutaneous (MEL888, MEL624) and one conjunctival (YUARGE 13-3064) melanoma cell line. Direct knockdown of EGR1 was accomplished using lentiviral vectors containing shRNA. Cell viability was measured using PrestoBlueHS Cell Viability Reagent. Total RNA and protein were assessed by qPCR and SimpleWestern. RNA-Seq demonstrated a profound reduction in EGR1 with MEK inhibitor treatment, prompting further study of melanoma cell lines. Following trametinib treatment of melanoma cells, viability was reduced in both cutaneous (MEL888 26%, P < 0.01; MEL624 27%, P < 0.001) and conjunctival (YUARGE 13-3064 33%, P < 0.01) melanoma compared with DMSO control, with confirmed EGR1 knockdown to 0.04-, 0.01-, and 0.16-fold DMSO-treated levels (all P < 0.05) in MEL888, MEL624, and YUARGE 13-3064, respectively. Targeted EGR1 knockdown using shRNA reduced viability in both cutaneous (MEL624 78%, P = 0.05) and conjunctival melanoma (YUARGE-13-3064 67%, P = 0.02). RNA-Sequencing in MEK inhibitor-treated cells identified EGR1 as a candidate effector molecule of interest. In a malignant melanoma cell population, MEK inhibition reduced viability in both cutaneous and conjunctival melanoma with a profound downstream reduction in EGR1 expression. Targeted knockdown of EGR1 reduced both cutaneous and conjunctival melanoma cell viability independent of MEK inhibition, suggesting a key role for EGR1 in melanoma pathobiology.
Collapse
Affiliation(s)
- David R Miley
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota
| | | | - Jose S Pulido
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota
- Wills Eye Hospital, Philadelphia, Pennsylvania
| | | | - Richard G Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Lauren A Dalvin
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
9
|
Fay CJ, Jakuboski S, Mclellan B, Allais BS, Semenov Y, Larocca CA, LeBoeuf NR. Diagnosis and Management of Dermatologic Adverse Events from Systemic Melanoma Therapies. Am J Clin Dermatol 2023; 24:765-785. [PMID: 37395930 PMCID: PMC10796164 DOI: 10.1007/s40257-023-00790-8] [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] [Accepted: 05/02/2023] [Indexed: 07/04/2023]
Abstract
The advent of protein kinase inhibitors and immunotherapy has profoundly improved the management of advanced melanoma. However, with these therapeutic advancements also come drug-related toxicities that have the potential to affect various organ systems. We review dermatologic adverse events from targeted (including BRAF and MEK inhibitor-related) and less commonly used melanoma treatments, with a focus on diagnosis and management. As immunotherapy-related toxicities have been extensively reviewed, herein, we discuss injectable talimogene laherparepvec and touch on recent breakthroughs in the immunotherapy space. Dermatologic adverse events may severely impact quality of life and are associated with response and survival. It is therefore essential that clinicians are aware of their diverse presentations and management strategies.
Collapse
Affiliation(s)
- Christopher J Fay
- Department of Dermatology, Brigham and Women's Hospital, and the Center for Cutaneous Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA, USA
| | | | - Beth Mclellan
- Department of Dermatology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Blair S Allais
- Department of Dermatology, Brigham and Women's Hospital, and the Center for Cutaneous Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Yevgeniy Semenov
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cecilia A Larocca
- Department of Dermatology, Brigham and Women's Hospital, and the Center for Cutaneous Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Nicole R LeBoeuf
- Department of Dermatology, Brigham and Women's Hospital, and the Center for Cutaneous Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Cao L, Chen S, Sun R, Ashby CR, Wei L, Huang Z, Chen ZS. Darovasertib, a novel treatment for metastatic uveal melanoma. Front Pharmacol 2023; 14:1232787. [PMID: 37576814 PMCID: PMC10419210 DOI: 10.3389/fphar.2023.1232787] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
The FDA granted orphan drug designation to darovasertib, a first-in-class oral, small molecular inhibitor of protein kinase C (PKC), for the treatment of uveal melanoma, on 2 May 2022. Primary uveal melanoma has a high risk of progressing to metastatic uveal melanoma, with a poor prognosis. The activation of the PKC and mitogen-activated protein kinase pathways play an essential role in the pathogenesis of uveal melanoma, and mutations in the G protein subunit alpha q (GNAQ), and G protein subunit alpha11 (GNA11) genes are considered early events in the development of uveal melanoma. Compared to other PKC inhibitors, such as sotrastaurin and enzastaurin, darovasertib is significantly more potent in inhibiting conventional (α, β) and novel (δ, ϵ, η, θ) PKC proteins and has a better tolerability and safety profile. Current Phase I/II clinical trials indicated that darovasertib, combined with the Mitogen-activated protein kinase/Extracellular (MEK) inhibitors, binimetinib or crizotinib, produced a synergistic effect of uveal melanoma. In this article, we summarize the development of drugs for treating uveal melanomas and discuss problems associated with current treatments. We also discuss the mechanism of action, pharmacokinetic profile, adverse effects, and clinical trial for darovasertib, and future research directions for treating uveal melanoma.
Collapse
Affiliation(s)
- Lei Cao
- Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Shuzhen Chen
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Rainie Sun
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
- Stuyvesant High School, New York, NY, United States
| | - Charles R. Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| | - Liuya Wei
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Zoufang Huang
- Ganzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| |
Collapse
|
11
|
Perez MC, Depalo DK, Zager JS. A safety review of recently approved and late-stage trial treatments for metastatic melanoma: systemic and regional therapies. Expert Opin Drug Saf 2023; 22:789-797. [PMID: 37551723 DOI: 10.1080/14740338.2023.2245333] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/30/2023] [Accepted: 08/03/2023] [Indexed: 08/09/2023]
Abstract
INTRODUCTION Advanced melanoma accounts for the majority of skin cancer-associated deaths. Over the past 15 years, there has been a dramatic change in the treatment options and prognosis for patients with advanced melanoma secondary to the development of novel systemic immunotherapies (IO) and targeted therapies. In addition to these novel systemic therapies, regional therapies (intralesional and perfusional) also continue to play a major role in the management of these patients. AREAS COVERED In this article, we review recent updates in the management of advanced melanoma via Medline (PubMed) and Google Scholar, including recently published trials in the metastatic, adjuvant, and neoadjuvant settings. We also review recently published trials for regional therapies and discuss future directions in the management of patients with advanced melanoma. EXPERT OPINION A significant portion of patients with advanced melanoma will develop recurrent or progressive disease following treatment with IO or targeted therapy. Therefore, identifying not only the appropriate therapeutic agent but also the sequence and duration of treatment is pivotal for these patients.
Collapse
Affiliation(s)
- Matthew C Perez
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa FL, United States of America
| | - Danielle K Depalo
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa FL, United States of America
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa FL, United States of America
| |
Collapse
|
12
|
Petzold A, Steeb T, Wessely A, Koch EAT, Vera J, Berking C, Heppt MV. Is tebentafusp superior to combined immune checkpoint blockade and other systemic treatments in metastatic uveal melanoma? A comparative efficacy analysis with population adjustment. Cancer Treat Rev 2023; 115:102543. [PMID: 36931146 DOI: 10.1016/j.ctrv.2023.102543] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/14/2023]
Abstract
BACKGROUND Distinct systemic treatments exist for metastatic uveal melanoma. Tebentafusp and combined immune checkpoint blockade (ICB) with ipilimumab plus anti-PD-1 antibodies are the most commonly used treatment options but their comparative efficacy is unclear. The aim of this study is to compare currently available systemic treatments regarding overall survival (OS) and progression-free survival (PFS) with a focus on the comparison of tebentafusp versus combined ICB. METHODS The protocol for this study was defined a priori and registered online in the PROSPERO international prospective register of systematic reviews (CRD42022308356, date of registration: 7.2.2022). We performed a systematic literature search in Medline, Embase, and Central to identify eligible studies reporting Kaplan-Meier curves or individual-level survival data showing OS and PFS for metastatic uveal melanoma patients treated with systemic treatments. Kaplan-Meier curves were digitized using the "WebPlotDigitizer" program. Individual-level survival data were subsequently remodelled and pooled for distinct treatment groups. To compare the OS of tebentafusp versus combined ICB, we used matching-adjusted indirect comparison (MAIC), two-stage MAIC (2SMAIC), and simulated treatment comparison (STC) together with digitized individual-level survival data as population-adjusted models. RESULTS Overall, 55 independent studies were included of which 2,682 patients were evaluable for OS and 2,258 for PFS. Tebentafusp showed the highest median OS (mOS) of 22.4 months (95% confidence interval (CI): 19.9-29.6) compared to combined ICB (mOS: 15.7 months (95% CI: 14.4-17.9)), anti-PD-(L)1 antibody (mOS: 10.9 months (95% CI: 9.8-13.4)), chemotherapy (mOS: 9.95 months (95% CI: 8.9-11.2)), targeted therapies (mOS: 8.86 months (95% CI: 7.5-10.8)), and anti-CTLA-4 antibody (mOS: 7.8 months (95% CI: 6.8-9.3). The median PFS (mPFS) was similar among the treatment groups ranging from 2.7 months to 3.4 months. For the comparison of tebentafusp versus combined ICB, the hazard ratio (HR) was 0.641 (95% CI: 0.449-0.915) in the unadjusted model, whereas the population-adjusted models showed a HR of 0.386 (95% CI: 0.236-0.631) using MAIC, 0.378 (95% CI: 0.234-0.612) applying 2SMAIC and 0.284 (95% CI: 0.184-0.440) using STC. CONCLUSIONS Tebentafusp achieved the best results compared to combined ICB and other systemic treatments, although these results have to be interpreted with caution due to the approximative methodical approach and high heterogeneity of included studies.
Collapse
Affiliation(s)
- Anne Petzold
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany; Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Uniklinikum Erlangen, Erlangen, Germany.
| | - Theresa Steeb
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany; Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Uniklinikum Erlangen, Erlangen, Germany.
| | - Anja Wessely
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany; Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Uniklinikum Erlangen, Erlangen, Germany.
| | - Elias A T Koch
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany; Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Uniklinikum Erlangen, Erlangen, Germany.
| | - Julio Vera
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany; Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Uniklinikum Erlangen, Erlangen, Germany.
| | - Carola Berking
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany; Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Uniklinikum Erlangen, Erlangen, Germany.
| | - Markus V Heppt
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany; Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Uniklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
13
|
Vogel A, Ochsenreither S, Zager JS, Wacker F, Saborowski A. Chemosaturation for primary and secondary liver malignancies: A comprehensive update of current evidence. Cancer Treat Rev 2023; 113:102501. [PMID: 36587472 DOI: 10.1016/j.ctrv.2022.102501] [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: 11/04/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/25/2022]
Abstract
Regional therapies for primary and secondary liver tumors have garnered interest in recent years and several types of treatment approaches have been pursued to control disease, palliate symptoms, and extend survival. Chemosaturation is an innovative way to deliver high-dose chemotherapy to the liver via the hepatic artery. Within the last decade, "isolated hepatic perfusion" (IHP) has evolved from an open surgical approach to a minimally invasive procedure, now termed "chemosaturation" (CS) with "percutaneous hepatic perfusion (PHP)". The most conclusive data on CS-PHP is currently available for patients with hepatic metastases from uveal melanoma (UM) - a rare but devastating disease with a poor long-term survival rate. A global phase-3 study and several cohort studies have provided compelling evidence that CS-PHP is an effective salvage treatment for liver-dominant metastatic UM in institutions with appropriate expertise. In this review we provide an overview on the technique, available clinical data, including safety and efficacy, and potential indications for CS-PHP.
Collapse
Affiliation(s)
- Arndt Vogel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.
| | - Sebastian Ochsenreither
- Department of Hematology, Oncology and Tumor Immunology, Charité Campus Benjamin Franklin, Berlin, Germany; Charité Comprehensive Cancer Center, Berlin, Germany
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States; Department of Oncological Sciences, University of South Florida, Morsani School of Medicine, Tampa, FL, United States
| | - Frank Wacker
- Department of Radiology, Hannover Medical School, Hannover, Germany
| | - Anna Saborowski
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
14
|
Aumiller JL, Wedegaertner PB. Disruption of the interaction between mutationally activated Gα q and Gβγ attenuates aberrant signaling. J Biol Chem 2023; 299:102880. [PMID: 36626984 PMCID: PMC9926304 DOI: 10.1016/j.jbc.2023.102880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/14/2022] [Accepted: 12/31/2022] [Indexed: 01/09/2023] Open
Abstract
Heterotrimeric G protein stimulation via G protein-coupled receptors promotes downstream proliferative signaling. Mutations can occur in Gα proteins which prevent GTP hydrolysis; this allows the G proteins to signal independently of G protein-coupled receptors and can result in various cancers, such as uveal melanoma (UM). Most UM cases harbor Q209L, Q209P, or R183C mutations in Gαq/11 proteins, rendering the proteins constitutively active (CA). Although it is generally thought that active, GTP-bound Gα subunits are dissociated from and signal independently of Gβγ, accumulating evidence indicates that some CA Gα mutants, such as Gαq/11, retain binding to Gβγ, and this interaction is necessary for signaling. Here, we demonstrate that disrupting the interaction between Gβγ and Gαq is sufficient to inhibit aberrant signaling driven by CA Gαq. Introduction of the I25A point mutation in the N-terminal α helical domain of CA Gαq to inhibit Gβγ binding, overexpression of the G protein Gαo to sequester Gβγ, and siRNA depletion of Gβ subunits inhibited or abolished CA Gαq signaling to the MAPK and YAP pathways. Moreover, in HEK 293 cells and in UM cell lines, we show that Gαq-Q209P and Gαq-R183C are more sensitive to the loss of Gβγ interaction than Gαq-Q209L. Our study challenges the idea that CA Gαq/11 signals independently of Gβγ and demonstrates differential sensitivity between the Gαq-Q209L, Gαq-Q209P, and Gαq-R183C mutants.
Collapse
|
15
|
Lapadula D, Lam B, Terai M, Sugase T, Tanaka R, Farias E, Kadamb R, Lopez-Anton M, Heine CC, Modasia B, Aguirre-Ghiso JA, Aplin AE, Sato T, Benovic JL. IGF1R Inhibition Enhances the Therapeutic Effects of Gq/11 Inhibition in Metastatic Uveal Melanoma Progression. Mol Cancer Ther 2023; 22:63-74. [PMID: 36223548 PMCID: PMC9812929 DOI: 10.1158/1535-7163.mct-22-0147] [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: 03/01/2022] [Revised: 08/04/2022] [Accepted: 10/06/2022] [Indexed: 02/03/2023]
Abstract
Uveal melanoma (UM) is the most common intraocular tumor in adults, and up to 50% of patients develop metastatic disease, which remains uncurable. Because patients with metastatic UM have an average survival of less than 1 year after diagnosis, there is an urgent need to develop new treatment strategies. Although activating mutations in Gαq or Gα11 proteins are major drivers of pathogenesis, the therapeutic intervention of downstream Gαq/11 targets has been unsuccessful in treating UM, possibly due to alternative signaling pathways and/or resistance mechanisms. Activation of the insulin-like growth factor 1 (IGF1) signaling pathway promotes cell growth, metastasis, and drug resistance in many types of cancers, including UM, where expression of the IGF1 receptor (IGF1R) correlates with a poor prognosis. In this article, we show that direct inhibition of Gαq/11 by the cyclic depsipeptide YM-254890 in combination with inhibition of IGF1R by linsitinib cooperatively inhibits downstream signaling and proliferation of UM cells. We further demonstrate that a 2-week combination treatment of 0.3 to 0.4 mg/kg of YM-254890 administered by intraperitoneal injection and 25 to 40 mg/kg linsitinib administered by oral gavage effectively inhibits the growth of metastatic UM tumors in immunodeficient NOD scid gamma (NSG) mice and identifies the IGF1 pathway as a potential resistance mechanism in response to Gαq/11 inhibition in UM. These data suggest that the combination of Gαq/11 and IGF1R inhibition provides a promising therapeutic strategy to treat metastatic UM.
Collapse
Affiliation(s)
- Dominic Lapadula
- Sidney Kimmel Cancer Center at Jefferson, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Bao Lam
- Thomas Jefferson University, Philadelphia, PA, United States
| | - Mizue Terai
- Thomas Jefferson University, Philadelphia, PA, United States
| | - Takahito Sugase
- Thomas Jefferson University, Philadelphia, PA, United States
| | - Ryota Tanaka
- Thomas Jefferson University, Philadelphia, PA, United States
| | - Eduardo Farias
- Icahn School of Medicine at Mount, New York, NY, United States
| | - Rama Kadamb
- Albert Einstein College of Medicine, Bronx, NewYork, United States
| | | | - Christian C Heine
- Sidney Kimmel Cancer Center at Jefferson, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | | | | | - Andrew E Aplin
- Thomas Jefferson University, Philadelphia, PA, United States
| | - Takami Sato
- Thomas Jefferson University, Philadelphia, PA, United States
| | - Jeffrey L Benovic
- Sidney Kimmel Cancer Center at Jefferson, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| |
Collapse
|
16
|
Zhang GM, Huang SS, Ye LX, Liu XL, Shi WH, Ren ZL, Zhou RH, Zhang JJ, Pan JX, Liu SW, Yu L, Li YL. Reciprocal positive regulation between BRD4 and YAP in GNAQ-mutant uveal melanoma cells confers sensitivity to BET inhibitors. Pharmacol Res 2022; 184:106464. [PMID: 36162600 DOI: 10.1016/j.phrs.2022.106464] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022]
Abstract
Uveal melanoma (UM) is the most common intraocular cancer in adults. UMs are usually initiated by a mutation in GNAQ or GNA11 (encoding Gq or G11, respectively), unlike cutaneous melanomas (CMs), which usually carry a BRAF or NRAS mutation. Currently, there are no clinically effective targeted therapies for UM carrying Gq/11 mutations. Here, we identified a causal link between Gq activating mutations and hypersensitivity to bromodomain and extra-terminal (BET) inhibitors. BET inhibitors transcriptionally repress YAP via BRD4 regardless of Gq mutation status, independently of Hippo core components LATS1/2. In contrast, YAP/TAZ downregulation reduces BRD4 transcription exclusively in Gq-mutant cells and LATS1/2 double knockout cells, both of which are featured by constitutively active YAP/TAZ. The transcriptional interdependency between BRD4 and YAP identified in Gq-mutated cells is responsible for the preferential inhibitory effect of BET inhibitors on the growth and dissemination of Gq-mutated UM cells compared to BRAF-mutated CM cells in both culture cells and animal models. Our findings suggest BRD4 as a viable therapeutic target for Gq-driven UMs that are addicted to unrestrained YAP function.
Collapse
Affiliation(s)
- Gui-Ming Zhang
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Si-Si Huang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lin-Xuan Ye
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Lian Liu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wen-Hui Shi
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhong-Lu Ren
- College of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Run-Hua Zhou
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jia-Jie Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jing-Xuan Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Shu-Wen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Le Yu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
| | - Yi-Lei Li
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
17
|
Zhang Y, Zhang B, Li Y, Dai Y, Li J, Li D, Xia Z, Zhang J, Liu P, Chen M, Jiao B, Ren R. Palmitoylation of GNAQ/11 is critical for tumor cell proliferation and survival in GNAQ/11-mutant uveal melanoma. Front Med 2022; 16:784-798. [PMID: 35997986 DOI: 10.1007/s11684-021-0911-0] [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: 06/01/2021] [Accepted: 11/25/2021] [Indexed: 11/04/2022]
Abstract
More than 85% of patients with uveal melanoma (UM) carry a GNAQ or GNA11 mutation at a hotspot codon (Q209) that encodes G protein α subunit q/11 polypeptides (Gαq/11). GNAQ/11 relies on palmitoylation for membrane association and signal transduction. Despite the palmitoylation of GNAQ/11 was discovered long before, its implication in UM remains unclear. Here, results of palmitoylation-targeted mutagenesis and chemical interference approaches revealed that the loss of GNAQ/11 palmitoylation substantially affected tumor cell proliferation and survival in UM cells. Palmitoylation inhibition through the mutation of palmitoylation sites suppressed GNAQ/11Q209L-induced malignant transformation in NIH3T3 cells. Importantly, the palmitoylation-deficient oncogenic GNAQ/11 failed to rescue the cell death initiated by the knock down of endogenous GNAQ/11 oncogenes in UM cells, which are much more dependent on Gαq/11 signaling for cell survival and proliferation than other melanoma cells without GNAQ/11 mutations. Furthermore, the palmitoylation inhibitor, 2-bromopalmitate, also specifically disrupted Gαq/11 downstream signaling by interfering with the MAPK pathway and BCL2 survival pathway in GNAQ/11-mutant UM cells and showed a notable synergistic effect when applied in combination with the BCL2 inhibitor, ABT-199, in vitro. The findings validate that GNAQ/11 palmitoylation plays a critical role in UM and may serve as a promising therapeutic target for GNAQ/11-driven UM.
Collapse
Affiliation(s)
- Yan Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Baoyuan Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yongyun Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200001, China
| | - Yuting Dai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiaoyang Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Donghe Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhizhou Xia
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jianming Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ping Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ming Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Bo Jiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Ruibao Ren
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Department of Biology, Brandeis University, Waltham, MA, 02454, USA.
| |
Collapse
|
18
|
Sundaramurthi H, Giricz Z, Kennedy BN. Evaluation of the Therapeutic Potential of Histone Deacetylase 6 Inhibitors for Primary and Metastatic Uveal Melanoma. Int J Mol Sci 2022; 23:ijms23169378. [PMID: 36012642 PMCID: PMC9409113 DOI: 10.3390/ijms23169378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/26/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Patients diagnosed with metastatic uveal melanoma (MUM) have a poor survival prognosis. Unfortunately for this rare disease, there is no known cure and suitable therapeutic options are limited. HDAC6 inhibitors (HDAC6i) are currently in clinical trials for other cancers and show potential beneficial effects against tumor cell survival in vitro and in vivo. In MUM cells, HDAC6i show an anti-proliferative effect in vitro and in preclinical xenograft models. The use of HDAC6 inhibitors as a treatment option for MUM should be explored further. Therefore, this review discusses (1) what is known about HDAC6i in MUM and (2) whether HDAC6 inhibitors offer a potential therapeutic option for MUM.
Collapse
Affiliation(s)
- Husvinee Sundaramurthi
- UCD Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
- UCD School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8 Dublin, Ireland
- Systems Biology Ireland, University College Dublin, D04 V1W8 Dublin, Ireland
- UCD School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Zoltán Giricz
- Pharmahungary Group, 6720 Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary
| | - Breandán N. Kennedy
- UCD Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
- UCD School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8 Dublin, Ireland
- Correspondence:
| |
Collapse
|
19
|
Li Y, Yang J, Zhang Q, Xu S, Sun W, Ge S, Xu X, Jager MJ, Jia R, Zhang J, Fan X. Copper ionophore elesclomol selectively targets GNAQ/11-mutant uveal melanoma. Oncogene 2022; 41:3539-3553. [PMID: 35697803 DOI: 10.1038/s41388-022-02364-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 05/18/2022] [Accepted: 05/26/2022] [Indexed: 01/10/2023]
Abstract
Unlike cutaneous melanoma, uveal melanoma (UM) is characterized by mutations in GNAQ and GNA11 and remains a fatal disease because there is essentially no effective targeted therapy or immunotherapy available. We report the discovery of the copper ionophore elesclomol as a GNAQ/11-specific UM inhibitor. Elesclomol was identified in a differential cytotoxicity screen of an in-house tool compound library, and its in vivo pharmacological efficacy was further confirmed in zebrafish and mouse UM models. Mechanistically, elesclomol transports copper to mitochondria and produces a large amount of reactive oxygen species (ROS) as Cu(II) is reduced to Cu(I) in GNAQ/11-mutant UM cells, which selectively activates LATS1 kinase in the Hippo signaling pathway and consequently promotes YAP phosphorylation and inhibits its nuclear accumulation. The inactivation of YAP downregulates the expression of SNAI2, which in turn suppresses the migration of UM cells. These findings were cross validated by our clinical observation that YAP activation was found specifically in UM samples with a GNAQ/11 mutation. Furthermore, addition of binimetinib, a MEK inhibitor, to elesclomol increased its synthetic lethality to GNAQ/11-mutant UM cells, thereby overriding drug resistance. This effect was confirmed in an orthotopic xenograft model and in a patient-derived xenograft model of UM. These studies reveal a novel mechanistic basis for repurposing elesclomol by showing that copper homeostasis is a GNAQ/11-specific vulnerability in UM. Elesclomol may provide a new therapeutic path for selectively targeting malignant GNAQ/11-mutant UM.
Collapse
Affiliation(s)
- Yongyun Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jie Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Qianqian Zhang
- National Research Center for Translational Medicine (Shanghai), State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shiqiong Xu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Wei Sun
- National Research Center for Translational Medicine (Shanghai), State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Jianming Zhang
- National Research Center for Translational Medicine (Shanghai), State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| |
Collapse
|
20
|
Leyvraz S, Konietschke F, Peuker C, Schütte M, Kessler T, Ochsenreither S, Ditzhaus M, Sprünken ED, Dörpholz G, Lamping M, Rieke DT, Klinghammer K, Burock S, Ulrich C, Poch G, Schäfer R, Klauschen F, Joussen A, Yaspo ML, Keilholz U. Biomarker-driven therapies for metastatic uveal melanoma: A prospective precision oncology feasibility study. Eur J Cancer 2022; 169:146-155. [DOI: 10.1016/j.ejca.2022.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 02/07/2023]
|
21
|
Glinkina K, Groenewoud A, Teunisse AFAS, Snaar-Jagalska BE, Jochemsen AG. Novel Treatments of Uveal Melanoma Identified with a Synthetic Lethal CRISPR/Cas9 Screen. Cancers (Basel) 2022; 14:3186. [PMID: 35804957 PMCID: PMC9264875 DOI: 10.3390/cancers14133186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/21/2022] [Accepted: 06/27/2022] [Indexed: 01/30/2023] Open
Abstract
Currently, no systemic treatment is approved as the standard of care for metastatic uveal melanoma (UM). mTOR has been evaluated as a drug target in UM. However, one of the main limitations is dose reduction due to adverse effects. The combination of everolimus with another targeted agent would allow the reduction of the dose of a single drug, thus widening the therapeutic window. In our study, we aimed to identify a synergistic combination with everolimus in order to develop a novel treatment option for metastatic UM. We exploited CRISPR-Cas9 synthetic lethality screening technology to search for an efficient combination. IGF1R and PRKDC and several other genes were identified as hits in the screen. We investigated the effect of the combination of everolimus with the inhibitors targeting IGF1R and DNA-PKcs on the survival of UM cell lines. These combinations synergistically slowed down cell growth but did not induce apoptosis in UM cell lines. These combinations were tested on PDX UM in an in vivo model, but we could not detect tumor regression. However, we could find significant activity of the dual DNA-PKcs/mTOR inhibitor CC-115 on PDX UM in the in vivo model.
Collapse
Affiliation(s)
- Kseniya Glinkina
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (K.G.); (A.F.A.S.T.)
| | - Arwin Groenewoud
- Department of Molecular Cell Biology, Institute of Biology, Leiden University, 2300 RC Leiden, The Netherlands; (A.G.); (B.E.S.-J.)
| | - Amina F. A. S. Teunisse
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (K.G.); (A.F.A.S.T.)
| | - B. Ewa Snaar-Jagalska
- Department of Molecular Cell Biology, Institute of Biology, Leiden University, 2300 RC Leiden, The Netherlands; (A.G.); (B.E.S.-J.)
| | - Aart G. Jochemsen
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (K.G.); (A.F.A.S.T.)
| |
Collapse
|
22
|
Phelps GB, Hagen HR, Amsterdam A, Lees JA. MITF deficiency accelerates GNAQ-driven uveal melanoma. Proc Natl Acad Sci U S A 2022; 119:e2107006119. [PMID: 35512098 PMCID: PMC9172632 DOI: 10.1073/pnas.2107006119] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 02/17/2022] [Indexed: 01/02/2023] Open
Abstract
Cutaneous melanoma (CM) and uveal melanoma (UM) both originate from the melanocytic lineage but are primarily driven by distinct oncogenic drivers, BRAF/NRAS or GNAQ/GNA11, respectively. The melanocytic master transcriptional regulator, MITF, is essential for both CM development and maintenance, but its role in UM is largely unexplored. Here, we use zebrafish models to dissect the key UM oncogenic signaling events and establish the role of MITF in UM tumors. Using a melanocytic lineage expression system, we showed that patient-derived mutations of GNAQ (GNAQQ209L) or its upstream CYSLTR2 receptor (CYSLTR2L129Q) both drive UM when combined with a cooperating mutation, tp53M214K/M214K. The tumor-initiating potential of the major GNAQ/11 effector pathways, YAP, and phospholipase C-β (PLCβ)–ERK was also investigated in this system and thus showed that while activated YAP (YAPAA) induced UM with high potency, the patient-derived PLCβ4 mutation (PLCB4D630Y) very rarely yielded UM tumors in the tp53M214K/M214K context. Remarkably, mitfa deficiency was profoundly UM promoting, dramatically accelerating the onset and progression of tumors induced by Tg(mitfa:GNAQQ209L);tp53M214K/M214K or Tg(mitfa:CYSLTR2L129Q);tp53M214K/M214K. Moreover, mitfa loss was sufficient to cooperate with GNAQQ209L to drive tp53–wild type UM development and allowed Tg(mitfa:PLCB4D630Y);tp53M214K/M214K melanocyte lineage cells to readily form tumors. Notably, all of the mitfa−/− UM tumors, including those arising in Tg(mitfa:PLCB4D630Y);tp53M214K/M214K;mitfa−/− zebrafish, displayed nuclear YAP while lacking hyperactive ERK indicative of PLCβ signaling. Collectively, these data show that YAP signaling is the major mediator of UM and that MITF acts as a bona fide tumor suppressor in UM in direct opposition to its essential role in CM.
Collapse
Affiliation(s)
- Grace B. Phelps
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Hannah R. Hagen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Adam Amsterdam
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Jacqueline A. Lees
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
23
|
Fei X, Xie X, Qin R, Wang A, Meng X, Sun F, Zhao Y, Jiang D, Chen H, Huang Q, Ji X, Wang Z. Proteomics analysis: inhibiting the expression of P62 protein by chloroquine combined with dacarbazine can reduce the malignant progression of uveal melanoma. BMC Cancer 2022; 22:408. [PMID: 35421957 PMCID: PMC9009011 DOI: 10.1186/s12885-022-09499-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 04/05/2022] [Indexed: 11/10/2022] Open
Abstract
Background Although uveal melanoma (UM) at the early stage is controllable to some extent, it inevitably ultimately leads to death due to its metastasis. At present, the difficulty is that there is no way to effectively tackle the metastasis. It is hypothesized that these will be treated by target molecules, but the recognized target molecule has not yet been found. In this study, the target molecule was explored through proteomics. Methods Transgenic enhanced green fluorescent protein (EGFP) inbred nude mice, which spontaneously display a tumor microenvironment (TME), were used as model animal carriers. The UM cell line 92.1 was inoculated into the brain ventricle stimulating metastatic growth of UM, and a graft re-cultured Next, the UM cell line 92.1-A was obtained through monoclonal amplification, and a differential proteomics database, between 92.1 and ectopic 92.1-A, was established. Finally, bioinformatics methodologies were adopted to optimize key regulatory proteins, and in vivo and in vitro functional verification and targeted drug screening were performed. Results Cells and tissues displaying green fluorescence in animal models were determined as TME characteristics provided by hosts. The data of various biological phenotypes detected proved that 92.1-A were more malignant than 92.1. Besides this malignancy, the key protein p62 (SQSTM1), selected from 5267 quantifiable differential proteomics databases, was a multifunctional autophagy linker protein, and its expression could be suppressed by chloroquine and dacarbazine. Inhibition of p62 could reduce the malignancy degree of 92.1-A. Conclusions As the carriers of human UM orthotopic and ectopic xenotransplantation, transgenic EGFP inbred nude mice clearly display the characteristics of TME. In addition, the p62 protein optimized by the proteomics is the key protein that increases the malignancy of 92.1 cells, which therefore provides a basis for further exploration of target molecule therapy for refractory metastatic UM. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09499-z.
Collapse
|
24
|
Uveal Melanoma Cell Line Proliferation Is Inhibited by Ricolinostat, a Histone Deacetylase Inhibitor. Cancers (Basel) 2022; 14:cancers14030782. [PMID: 35159049 PMCID: PMC8833954 DOI: 10.3390/cancers14030782] [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: 12/16/2021] [Revised: 01/19/2022] [Accepted: 01/30/2022] [Indexed: 12/26/2022] Open
Abstract
Metastatic uveal melanoma (MUM) is characterized by poor patient survival. Unfortunately, current treatment options demonstrate limited benefits. In this study, we evaluate the efficacy of ACY-1215, a histone deacetylase inhibitor (HDACi), to attenuate growth of primary ocular UM cell lines and, in particular, a liver MUM cell line in vitro and in vivo, and elucidate the underlying molecular mechanisms. A significant (p = 0.0001) dose-dependent reduction in surviving clones of the primary ocular UM cells, Mel270, was observed upon treatment with increasing doses of ACY-1215. Treatment of OMM2.5 MUM cells with ACY-1215 resulted in a significant (p = 0.0001), dose-dependent reduction in cell survival and proliferation in vitro, and in vivo attenuation of primary OMM2.5 xenografts in zebrafish larvae. Furthermore, flow cytometry revealed that ACY-1215 significantly arrested the OMM2.5 cell cycle in S phase (p = 0.0001) following 24 h of treatment, and significant apoptosis was triggered in a time- and dose-dependent manner (p < 0.0001). Additionally, ACY-1215 treatment resulted in a significant reduction in OMM2.5 p-ERK expression levels. Through proteome profiling, the attenuation of the microphthalmia-associated transcription factor (MITF) signaling pathway was linked to the observed anti-cancer effects of ACY-1215. In agreement, pharmacological inhibition of MITF signaling with ML329 significantly reduced OMM2.5 cell survival and viability in vitro (p = 0.0001) and reduced OMM2.5 cells in vivo (p = 0.0006). Our findings provide evidence that ACY-1215 and ML329 are efficacious against growth and survival of OMM2.5 MUM cells.
Collapse
|
25
|
Combination of Immune Checkpoint Inhibitors and Liver-Specific Therapies in Liver-Metastatic Uveal Melanoma: Can We Thus Overcome Its High Resistance? Cancers (Basel) 2021; 13:cancers13246390. [PMID: 34945010 PMCID: PMC8699813 DOI: 10.3390/cancers13246390] [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: 11/01/2021] [Revised: 12/11/2021] [Accepted: 12/15/2021] [Indexed: 01/10/2023] Open
Abstract
Uveal Melanoma (UM) is a rare disease; however, it is the most common primary intraocular malignant tumor in adults. Hematogenous metastasis, occurring in up to 50% of cases, mainly to the liver (90%), is associated with poor clinical course and treatment failure. In contrast to dramatic benefits of immunotherapy in many tumor entities, as seen in cutaneous melanoma, immune checkpoint inhibitors (ICI) do not achieve comparable results in Metastatic UM (MUM). The aim of this study was to investigate whether the combination of ICI with liver-directed therapies provides a potential survival benefit for those affected. This retrospective, single-center study, including n = 45 patients with MUM, compared the effect of combining ICI with liver-directed therapy ("Cohort 1") with respect to standard therapies ("Cohort 2") on overall survival (OS). Our results revealed a significant survival difference between Cohort 1 (median OS 22.5 months) and Cohort 2 (median OS 11.4 months), indicating that this combination may enhance the efficacy of immunotherapy and thus provide a survival benefit. There is an urgent need for randomized, prospective trials addressing the combination of liver-directed therapies and various strategies of immunotherapy (such as ICI; IMCgp100; personalized vaccines) in order to establish regimens which finally improve the prognosis of patients with MUM.
Collapse
|
26
|
Lapadula D, Benovic JL. Targeting Oncogenic Gα q/11 in Uveal Melanoma. Cancers (Basel) 2021; 13:6195. [PMID: 34944815 PMCID: PMC8699590 DOI: 10.3390/cancers13246195] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 11/17/2022] Open
Abstract
Uveal melanoma is the most common intraocular cancer in adults and arises from the transformation of melanocytes in the uveal tract. While treatment of the primary tumor is often effective, 36-50% of patients develop metastatic disease primarily to the liver. While various strategies have been used to treat the metastatic disease, there remain no effective treatments that improve survival. Significant insight has been gained into the pathways that are altered in uveal melanoma, with mutually exclusive activating mutations in the GNAQ and GNA11 genes being found in over 90% of patients. These genes encode the alpha subunits of the hetetrotrimeric G proteins, Gq and G11, and mutations result in activation of several important signaling pathways, including phospholipase C and activation of the transcription factor YAP. In this review, we discuss current efforts to target various signaling pathways in the treatment of uveal melanoma including recent efforts to target Gq and G11 in mouse models. While selective targeting of Gq and G11 provides a potential therapeutic strategy to treat uveal melanoma, it is evident that improved inhibitors and methods of delivery are needed.
Collapse
Affiliation(s)
| | - Jeffrey L. Benovic
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| |
Collapse
|
27
|
Seedor RS, Orloff M, Sato T. Genetic Landscape and Emerging Therapies in Uveal Melanoma. Cancers (Basel) 2021; 13:5503. [PMID: 34771666 PMCID: PMC8582814 DOI: 10.3390/cancers13215503] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/26/2021] [Accepted: 10/30/2021] [Indexed: 12/12/2022] Open
Abstract
Despite successful treatment of primary uveal melanoma, up to 50% of patients will develop systemic metastasis. Metastatic disease portends a poor outcome, and no adjuvant or metastatic therapy has been FDA approved. The genetic landscape of uveal melanoma is unique, providing prognostic and potentially therapeutic insight. In this review, we discuss our current understanding of the molecular and cytogenetic mutations in uveal melanoma, and the importance of obtaining such information. Most of our knowledge is based on primary uveal melanoma and a better understanding of the mutational landscape in metastatic uveal melanoma is needed. Clinical trials targeting certain mutations such as GNAQ/GNA11, BAP1, and SF3B1 are ongoing and promising. We also discuss the role of liquid biopsies in uveal melanoma in this review.
Collapse
Affiliation(s)
- Rino S. Seedor
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (M.O.); (T.S.)
| | | | | |
Collapse
|
28
|
Potential of miRNA-Based Nanotherapeutics for Uveal Melanoma. Cancers (Basel) 2021; 13:cancers13205192. [PMID: 34680340 PMCID: PMC8534265 DOI: 10.3390/cancers13205192] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Human uveal melanoma (UM) is the most common primary intraocular tumor with high metastatic risk in adults. Currently, no effective treatment is available for metastatic UM; therefore, new therapeutic approaches are needed to improve overall survival. Given the increased understanding of microRNAs (miRNAs) and their roles in UM tumorigenesis and metastasis, miRNA-based therapy may offer the hope of improving therapeutic outcomes. This review summarizes the actions of select miRNAs examined in preclinical studies using miRNAs as therapeutic targets in UM. The focus of this review is the application of established nanotechnology-assisted delivery systems to overcome the limitations of therapeutic miRNAs. A blend of therapeutic miRNAs and nanodelivery systems may facilitate the translation of miRNA therapies to clinical settings. Abstract Uveal melanoma (UM) is the most common adult intraocular cancer, and metastatic UM remains deadly and incurable. UM is a complex disease associated with the deregulation of numerous genes and redundant intracellular signaling pathways. As understanding of epigenetic dysregulation in the oncogenesis of UM has increased, the abnormal expression of microRNAs (miRNAs) has been found to be an epigenetic mechanism underlying UM tumorigenesis. A growing number of miRNAs are being found to be associated with aberrant signaling pathways in UM, and some have been investigated and functionally characterized in preclinical settings. This review summarizes the miRNAs with promising therapeutic potential for UM treatment, paying special attention to the therapeutic miRNAs (miRNA mimics or inhibitors) used to restore dysregulated miRNAs to their normal levels. However, several physical and physiological limitations associated with therapeutic miRNAs have prevented their translation to cancer therapeutics. With the advent of nanotechnology delivery systems, the development of effective targeted therapies for patients with UM has received great attention. Therefore, this review provides an overview of the use of nanotechnology drug delivery systems, particularly nanocarriers that can be loaded with therapeutic miRNAs for effective delivery into target cells. The development of miRNA-based therapeutics with nanotechnology-based delivery systems may overcome the barriers of therapeutic miRNAs, thereby enabling their translation to therapeutics, enabling more effective targeting of UM cells and consequently improving therapeutic outcomes.
Collapse
|
29
|
Nivolumab for patients with metastatic uveal melanoma previously untreated with ipilimumab: a single-institution retrospective study. Melanoma Res 2020; 30:76-84. [PMID: 31095037 DOI: 10.1097/cmr.0000000000000617] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We evaluated the efficacy of nivolumab in patients with metastatic uveal melanoma previously untreated with ipilimumab. We performed a retrospective study at the National Cancer Center Hospital in Tokyo, Japan, where nivolumab was approved 1 year earlier than ipilimumab. Clinical efficacy outcomes were determined by assessing best overall response according to the Response Evaluation Criteria in Solid Tumors (version 1.1), progression-free survival and overall survival. Fourteen patients were analyzed; none had received any prior systemic therapies although eight had undergone transarterial chemoembolization. The median follow-up period was 15 months. The objective response and disease control rates were 7.1% and 42.9%, respectively (one partial response and five stable diseases). The median progression-free survival and overall survival were 10 (range, 4-105) and 60 (range, 5-105) weeks, respectively. Liver metastases in three patients were all programmed cell death-1 ligand negative. Lower lactate dehydrogenase, development of vitiligo, and a neutrophil-to-lymphocyte ratio less than 5 at week 6 were associated with favorable progression-free survival and overall survival; of these, only a neutrophil-to-lymphocyte ratio less than 5 at week 6 was statistically significant. Even with the use of nivolumab before ipilimumab, metastatic uveal melanoma appears to remain refractory to nivolumab monotherapy. However, because one patient in our cohort achieved an objective response, and the median overall survival exceeded 1 year, treatment strategies that incorporate anti-PD1 antibody should be further investigated. Whether a neutrophil-to-lymphocyte ratio less than 5 at week 6 is a favorable early on-treatment marker should be validated in larger cohorts.
Collapse
|
30
|
Role of Natural Killer Cells in Uveal Melanoma. Cancers (Basel) 2020; 12:cancers12123694. [PMID: 33317028 PMCID: PMC7764114 DOI: 10.3390/cancers12123694] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Metastatic Uveal Melanoma (MUM) is a lethal malignancy with no durable treatment available to date. A vast majority of patients with MUM present with liver metastasis. The liver harbors metastatic disease with an apparent lack of a cytotoxic T cell response. It is becoming evident that MUM is not an immunologically silent malignancy and the investigation of non-T cell anti-tumor immunity is warranted. In this review, we highlight the relevance of Natural Killer (NK) cells in the biology and treatment of MUM. Potent anti-NK cell immunosuppression employed by uveal melanoma alludes to its vulnerability to NK cell cytotoxicity. On the contrary, micro-metastasis in the liver survive for several years within close vicinity of a plethora of circulating and liver-resident NK cells. This review provides unique perspectives into the potential role of NK cells in control or progression of uveal melanoma. Abstract Uveal melanoma has a high mortality rate following metastasis to the liver. Despite advances in systemic immune therapy, treatment of metastatic uveal melanoma (MUM) has failed to achieve long term durable responses. Barriers to success with immune therapy include the immune regulatory nature of uveal melanoma as well as the immune tolerant environment of the liver. To adequately harness the anti-tumor potential of the immune system, non-T cell-based approaches need to be explored. Natural Killer (NK) cells possess potent ability to target tumor cells via innate and adaptive responses. In this review, we discuss evidence that highlights the role of NK cell surveillance and targeting of uveal melanoma. We also discuss the repertoire of intra-hepatic NK cells. The human liver has a vast and diverse lymphoid population and NK cells comprise 50% of the hepatic lymphocytes. Hepatic NK cells share a common niche with uveal melanoma micro-metastasis within the liver sinusoids. It is, therefore, crucial to understand and investigate the role of intra-hepatic NK cells in the control or progression of MUM.
Collapse
|
31
|
Steeb T, Wessely A, Alter M, Bayerl C, Bender A, Bruning G, Dabrowski E, Debus D, Devereux N, Dippel E, Drexler K, Dücker P, Dummer R, Emmert S, Elsner P, Enk A, Gebhardt C, Gesierich A, Goebeler M, Goerdt S, Goetze S, Gutzmer R, Haferkamp S, Hansel G, Hassel JC, Heinzerling L, Kähler KC, Kaume KM, Krapf W, Kreuzberg N, Lehmann P, Livingstone E, Löffler H, Loquai C, Mauch C, Mangana J, Meier F, Meissner M, Moritz RKC, Maul LV, Müller V, Mohr P, Navarini A, Van Nguyen A, Pfeiffer C, Pföhler C, Posch C, Richtig E, Rompel R, Sachse MM, Sauder S, Schadendorf D, Schatton K, Schulze HJ, Schultz E, Schilling B, Schmuth M, Simon JC, Streit M, Terheyden P, Thiem A, Tüting T, Welzel J, Weyandt G, Wesselmann U, Wollina U, Ziemer M, Zimmer L, Zutt M, Berking C, Schlaak M, Heppt MV. Patterns of care and follow-up care of patients with uveal melanoma in German-speaking countries: a multinational survey of the German Dermatologic Cooperative Oncology Group (DeCOG). J Cancer Res Clin Oncol 2020; 147:1763-1771. [PMID: 33219855 PMCID: PMC8076157 DOI: 10.1007/s00432-020-03450-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 10/31/2020] [Indexed: 12/22/2022]
Abstract
Purpose Uveal melanoma (UM) is an orphan cancer of high unmet medical need. Current patterns of care and surveillance remain unclear as they are situated in an interdisciplinary setting. Methods A questionnaire addressing the patterns of care and surveillance in the management of patients with uveal melanoma was distributed to 70 skin cancer centers in Austria, Germany and Switzerland. Frequency distributions of responses for each item of the questionnaire were calculated. Results 44 of 70 (62.9%) skin cancer centers completed the questionnaire. Thirty-nine hospitals were located in Germany (88.6%), three in Switzerland (6.8%) and two in Austria (4.5%). The majority (68.2%) represented university hospitals. Most patients with metastatic disease were treated in certified skin cancer centers (70.7%, 29/41). Besides, the majority of patients with UM were referred to the respective skin cancer center by ophthalmologists (87.2%, 34/39). Treatment and organization of follow-up of patients varied across the different centers. 35.1% (14/37) of the centers stated to not perform any screening measures. Conclusion Treatment patterns of patients with uveal melanoma in Germany, Austria and Switzerland remain extremely heterogeneous. A guideline for the treatment and surveillance is urgently needed.
Collapse
Affiliation(s)
- Theresa Steeb
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany.,Comprehensive Cancer Center Erlangen, European Metropolitan Region of Nuremberg, Erlangen, Germany
| | - Anja Wessely
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany.,Comprehensive Cancer Center Erlangen, European Metropolitan Region of Nuremberg, Erlangen, Germany
| | - Mareike Alter
- Department of Dermatology, University Medical Center, Magdeburg, Germany
| | - Christiane Bayerl
- Department of Dermatology and Allergology, Skin Cancer Center Wiesbaden, Helios Dr. Horst Schmidt Clinics, Wiesbaden, Germany
| | - Armin Bender
- Department of Dermatology and Allergology, Philipps University Marburg, Marburg, Germany
| | - Guido Bruning
- Center for Venous and Dermatosurgery, Tabea Krankenhaus Hamburg, Hamburg, Germany
| | - Evelyn Dabrowski
- Department of Dermatology, Ludwigshafen Medical Center, Ludwigshafen, Germany
| | - Dirk Debus
- Department of Dermatology, Paracelsus Medical University Nuremberg, City Hospital of Nuremberg, Nuremberg, Germany
| | - Nina Devereux
- Center for Venous and Dermatosurgery, Tabea Krankenhaus Hamburg, Hamburg, Germany
| | - Edgar Dippel
- Department of Dermatology, Ludwigshafen Medical Center, Ludwigshafen, Germany
| | - Konstantin Drexler
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Pia Dücker
- Department of Dermatology, Hospital of Dortmund, Dortmund, Germany
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, University Zurich, Zürich, Switzerland
| | - Steffen Emmert
- Department of Dermatology and Venereology, University Medical Center, Rostock, Germany
| | - Peter Elsner
- Department of Dermatology, University Hospital, Jena, Germany
| | - Alexander Enk
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Christoffer Gebhardt
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anja Gesierich
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Matthias Goebeler
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Sergij Goerdt
- Department of Dermatology, Venerology and Allergology, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Steven Goetze
- Department of Dermatology, University Hospital, Jena, Germany
| | - Ralf Gutzmer
- Department of Dermatology and Allergy, Hannover Medical School, Skin Cancer Center Hannover, Hannover, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Gesina Hansel
- Department of Dermatology and Allergology, Städtisches Klinikum Dresden, Academic Teaching Hospital of the Technical University of Dresden, Dresden, Germany
| | - Jessica C Hassel
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Lucie Heinzerling
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany.,Comprehensive Cancer Center Erlangen, European Metropolitan Region of Nuremberg, Erlangen, Germany.,Department of Dermatology and Allergy, University Hospital, Ludwig-Maximilians-University (LMU), Frauenlobstr. 9-11, 80337, Munich, Germany
| | - Katharina C Kähler
- Department of Dermatology, Venereology and Allergology, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Kjell M Kaume
- Department of Dermatology and Allergology, Klinikum Bremen-Mitte, Bremen, Germany
| | - Wolfgang Krapf
- Department of Dermatology, SLK Hospital Heilbronn, Heilbronn, Germany
| | - Nicole Kreuzberg
- Department of Dermatology and Venerology, University of Cologne, Cologne, Germany
| | - Percy Lehmann
- Department of Dermatology, Helios University Hospital Wuppertal, Wuppertal, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Harald Löffler
- Department of Dermatology, SLK Hospital Heilbronn, Heilbronn, Germany
| | - Carmen Loquai
- Department of Dermatology, University Medical Center Mainz, Mainz, Germany
| | - Cornelia Mauch
- Department of Dermatology and Venerology, University of Cologne, Cologne, Germany
| | - Johanna Mangana
- Department of Dermatology, University Hospital Zurich, University Zurich, Zürich, Switzerland
| | - Friedegund Meier
- Department of Dermatology, University Hospital Dresden, Dresden, Germany
| | - Markus Meissner
- Department of Dermatology, Venereology and Allergology, Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - Rose K C Moritz
- Department of Dermatology and Venereology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Lara Valeska Maul
- Department of Dermatology, University Hospital Basel, Basel, Switzerland
| | - Verena Müller
- Department of Dermatology, Venerology and Allergology, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Peter Mohr
- Department of Dermatology, Elbe-Kliniken, Buxtehude, Germany
| | - Alexander Navarini
- Department of Dermatology, University Hospital Basel, Basel, Switzerland
| | - Ahn Van Nguyen
- Department of Dermatology, Venerology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christiane Pfeiffer
- Department of Dermatology and Allergology, University Hospital Augsburg, Augsburg, Germany.,Department of Dermatology and Allergology, University Hospital Ulm, Ulm, Germany
| | - Claudia Pföhler
- Department of Dermatology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Christian Posch
- Department of Dermatology and Allergy, School of Medicine, German Cancer Consortium (DKTK), Technical University of Munich, Munich, Germany
| | - Erika Richtig
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Rainer Rompel
- Department of Dermatology, Klinikum Kassel, Kassel, Germany
| | - Michael M Sachse
- Skin Cancer Center, Department of Dermatology, Allergology and Phlebology, Bremerhaven Reinkenheide, Bremerhaven, Germany
| | | | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kerstin Schatton
- Medical Faculty, Department of Dermatology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Hans-Joachim Schulze
- Department of Dermatology and Dermato-Histo-Pathology, Fachklinik Hornheide, Skin Cancer Centre, Münster, Germany
| | - Erwin Schultz
- Department of Dermatology, Paracelsus Medical University Nuremberg, City Hospital of Nuremberg, Nuremberg, Germany
| | - Bastian Schilling
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Matthias Schmuth
- Department of Dermatology, Venerology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jan C Simon
- Department of Dermatology, Venereology and Allergology, University Medical Center Leipzig, Leipzig, Germany
| | - Markus Streit
- Department of Dermatology, Hospital Aarau, Aarau, Switzerland
| | | | - Alexander Thiem
- Department of Dermatology and Venereology, University Medical Center, Rostock, Germany
| | - Thomas Tüting
- Department of Dermatology, University Medical Center, Magdeburg, Germany
| | - Julia Welzel
- Department of Dermatology and Allergology, University Hospital Augsburg, Augsburg, Germany
| | - Gerhard Weyandt
- Department of Dermatology and Allergology, Hospital Bayreuth, Bayreuth, Germany
| | - Ulrich Wesselmann
- Department of Dermatology, Helios University Hospital Wuppertal, Wuppertal, Germany
| | - Uwe Wollina
- Department of Dermatology and Allergology, Städtisches Klinikum Dresden, Academic Teaching Hospital of the Technical University of Dresden, Dresden, Germany
| | - Mirjana Ziemer
- Department of Dermatology, Venereology and Allergology, University Medical Center Leipzig, Leipzig, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Markus Zutt
- Department of Dermatology and Allergology, Klinikum Bremen-Mitte, Bremen, Germany
| | - Carola Berking
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany.,Comprehensive Cancer Center Erlangen, European Metropolitan Region of Nuremberg, Erlangen, Germany
| | - Max Schlaak
- Department of Dermatology and Allergy, University Hospital, Ludwig-Maximilians-University (LMU), Frauenlobstr. 9-11, 80337, Munich, Germany.
| | - Markus V Heppt
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany.,Comprehensive Cancer Center Erlangen, European Metropolitan Region of Nuremberg, Erlangen, Germany
| | | |
Collapse
|
32
|
Porcelli L, Mazzotta A, Garofoli M, Di Fonte R, Guida G, Guida M, Tommasi S, Azzariti A. Active notch protects MAPK activated melanoma cell lines from MEK inhibitor cobimetinib. Biomed Pharmacother 2020; 133:111006. [PMID: 33202284 DOI: 10.1016/j.biopha.2020.111006] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 10/26/2020] [Accepted: 11/08/2020] [Indexed: 12/18/2022] Open
Abstract
The crosstalk between Notch and MAPK pathway plays a role in MEK inhibitor resistance in BRAFV600E metastatic melanoma (MM) and promotes migration in GNAQQ209L uveal melanoma (UM) cells. We determined the cytotoxicity of combinatorial inhibition of MEK and Notch by cobimetinib and γ-secretase inhibitor (GSI) nirogacestat, in BRAFV600E and BRAF wt MM and GNAQQ209L UM cells displaying different Erk1/2 and Notch activation status, with the aim to elucidate the impact of Notch signaling in the response to MEK inhibitor. Overall the combination was synergic in BRAFV600E MM and GNAQQ209L UM cells and antagonistic in BRAF wt one. Focusing on UM cells, we found that cobimetinib resulted in G0/G1 phase arrest and apoptosis induction, whereas the combination with GSI increased treatment efficacy by inducing a senescent-like state of cells and by blocking migration towards liver cancer cells. Mechanistically, this was reflected in a strong reduction of cyclin D1, in the inactivation of retinoblastoma protein and in the increase of p27KIP1 expression levels. Of note, each drug alone prevented Notch signaling activation resulting in inhibition of c-jun(Ser63) and Hes-1 expression. The combination achieved the strongest inhibition on Notch signaling and on both c-jun(Ser63) and Erk1/2 activation level. In conclusion we unveiled a coordinate action of MAPK and Notch signaling in promoting proliferation of BRAFV600E MM and GNAQQ209L UM cells. Remarkably, the simultaneous inhibition of MEK and Notch signaling highlighted a role for the second pathway in protecting cells against senescence in GNAQQ209L UM cells treated with the MEK inhibitor.
Collapse
Affiliation(s)
- Letizia Porcelli
- Experimental Pharmacology Laboratory, Italia, 70124, Bari, Italy
| | | | | | - Roberta Di Fonte
- Experimental Pharmacology Laboratory, Italia, 70124, Bari, Italy
| | - Gabriella Guida
- Department of Basic Medical Sciences Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare 11, 70124 Bari, Italy
| | | | - Stefania Tommasi
- Molecular Diagnostics and Pharmacogenetics Unit IRCCS Istituto Tumori "Giovanni Paolo II" di Bari, Italia, 70124, Bari, Italy
| | - Amalia Azzariti
- Experimental Pharmacology Laboratory, Italia, 70124, Bari, Italy.
| |
Collapse
|
33
|
Li Y, Shi J, Yang J, Ge S, Zhang J, Jia R, Fan X. Uveal melanoma: progress in molecular biology and therapeutics. Ther Adv Med Oncol 2020; 12:1758835920965852. [PMID: 33149769 PMCID: PMC7586035 DOI: 10.1177/1758835920965852] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Uveal melanoma (UM) is the most common intraocular malignancy in adults. So far, no systemic therapy or standard treatment exists to reduce the risk of metastasis and improve overall survival of patients. With the increased knowledge regarding the molecular pathways that underlie the oncogenesis of UM, it is expected that novel therapeutic approaches will be available to conquer this disease. This review provides a summary of the current knowledge of, and progress made in understanding, the pathogenesis, genetic mutations, epigenetics, and immunology of UM. With the advent of the omics era, multi-dimensional big data are publicly available, providing an innovation platform to develop effective targeted and personalized therapeutics for UM patients. Indeed, recently, a great number of therapies have been reported specifically for UM caused by oncogenic mutations, as well as other etiologies. In this review, special attention is directed to advancements in targeted therapies. In particular, we discuss the possibilities of targeting: GNAQ/GNA11, PLCβ, and CYSLTR2 mutants; regulators of G-protein signaling; the secondary messenger adenosine diphosphate (ADP)-ribosylation factor 6 (ARF6); downstream pathways, such as those involving mitogen-activated protein kinase/MEK/extracellular signal-related kinase, protein kinase C (PKC), phosphoinositide 3-kinase/Akt/mammalian target of rapamycin (mTOR), Trio/Rho/Rac/Yes-associated protein, and inactivated BAP1; and immune-checkpoint proteins cytotoxic T-lymphocyte antigen 4 and programmed cell-death protein 1/programmed cell-death ligand 1. Furthermore, we conducted a survey of completed and ongoing clinical trials applying targeted and immune therapies for UM. Although drug combination therapy based on the signaling pathways involved in UM has made great progress, targeted therapy is still an unmet medical need.
Collapse
Affiliation(s)
- Yongyun Li
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jiahao Shi
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jie Yang
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jianming Zhang
- National Research Center for Translational Medicine, Shanghai State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Huangpu District, Shanghai 200001, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200001, China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Huangpu District, Shanghai 200001, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, 833 Zhizaoju Road, Huangpu District, Shanghai 200001, China
| |
Collapse
|
34
|
Truong A, Yoo JH, Scherzer MT, Sanchez JMS, Dale KJ, Kinsey CG, Richards JR, Shin D, Ghazi PC, Onken MD, Blumer KJ, Odelberg SJ, McMahon M. Chloroquine Sensitizes GNAQ/11-mutated Melanoma to MEK1/2 Inhibition. Clin Cancer Res 2020; 26:6374-6386. [PMID: 32933997 DOI: 10.1158/1078-0432.ccr-20-1675] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/03/2020] [Accepted: 09/11/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE Mutational activation of GNAQ or GNA11 (GNAQ/11), detected in >90% of uveal melanomas, leads to constitutive activation of oncogenic pathways, including MAPK and YAP. To date, chemo- or pathway-targeted therapies, either alone or in combination, have proven ineffective in the treatment of patients with metastatic uveal melanoma. EXPERIMENTAL DESIGN We tested the efficacy of chloroquine or hydroxychloroquine, in combination with MAPK pathway inhibition in GNAQ/11-mutated cells in vitro and in vivo and identified mechanisms of MEK1/2 inhibitor plus chloroquine-induced cytotoxicity. RESULTS Inhibition of GNAQ/11-mediated activation of MAPK signaling resulted in the induction of autophagy. Combined inhibition of Gα and autophagy or lysosome function resulted in enhanced cell death. Moreover, the combination of MEK1/2 inhibition, using trametinib, with the lysosome inhibitor, chloroquine, also increased cytotoxicity. Treatment of mice bearing GNAQ/11-driven melanomas with trametinib plus hydroxychloroquine resulted in inhibition of tumor growth and significantly prolonged survival. Interestingly, lysosomal- and autophagy-specific inhibition with bafilomycin A1 was not sufficient to promote cytotoxicity in combination with trametinib. However, the addition of YAP inhibition with trametinib plus bafilomycin A1 resulted in cell death at comparable levels to trametinib plus chloroquine (T/CQ) treatment. Furthermore, T/CQ-treated cells displayed decreased YAP nuclear localization and decreased YAP transcriptional activity. Expression of a constitutively active YAP5SA mutant conferred resistance to T/CQ-induced cell death. CONCLUSIONS These results suggest that YAP, MEK1/2, and lysosome function are necessary and critical targets for the therapy of GNAQ/11-driven melanoma, and identify trametinib plus hydroxychloroquine as a potential treatment strategy for metastatic uveal melanoma.
Collapse
Affiliation(s)
- Amanda Truong
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Jae Hyuk Yoo
- Molecular Medicine Program, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Michael T Scherzer
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | | | - Kali J Dale
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Conan G Kinsey
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Jackson R Richards
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah
- Molecular Medicine Program, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Donghan Shin
- Molecular Medicine Program, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Phaedra C Ghazi
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Michael D Onken
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri
| | - Kendall J Blumer
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Shannon J Odelberg
- Molecular Medicine Program, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Martin McMahon
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah.
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
- Department of Dermatology, University of Utah, Salt Lake City, Utah
| |
Collapse
|
35
|
MicroRNAs and Uveal Melanoma: Understanding the Diverse Role of These Small Molecular Regulators. Int J Mol Sci 2020; 21:ijms21165648. [PMID: 32781746 PMCID: PMC7460624 DOI: 10.3390/ijms21165648] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/31/2020] [Accepted: 08/04/2020] [Indexed: 12/12/2022] Open
Abstract
Uveal melanoma (UM) is a rare tumour of the eye, characterised by a high propensity to metastasise in half of all patients, most frequently to the liver. Although there are effective treatment options for the primary tumour, once metastasis has occurred prognosis is poor, with overall survival limited to months. Currently, there are no effective treatments for metastatic UM, despite the tumour having a well-defined signalling pathway to which many therapies have been directed. In an effort to develop novel treatment approaches, understanding the role of other signalling molecules, such as microRNAs, is fundamental. MicroRNAs (miRNAs) are small non-coding RNA molecules involved in posttranscriptional gene regulation, resulting in reduced target gene expression and subsequent protein translation. In UM, several dysregulated miRNAs have been proposed to play a functional role in disease progression, whereas others have been put forward as clinical biomarkers of high-risk disease following isolation from blood, plasma and exosomes. Most recently, analyses of large datasets have identified promising prognostic miRNA signatures and panels. This review navigates the plethora of aberrant miRNAs disclosed so far in UM, and maps these to signalling pathways, which could be targeted in future therapies for the disseminated disease.
Collapse
|
36
|
Thornton S, Kalirai H, Aughton K, Coupland SE. Unpacking the genetic etiology of uveal melanoma. EXPERT REVIEW OF OPHTHALMOLOGY 2020. [DOI: 10.1080/17469899.2020.1785872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Sophie Thornton
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Liverpool Clinical Laboratories, Liverpool University Hospitals Foundation Trusts, Liverpool, UK
| | - Helen Kalirai
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Liverpool Clinical Laboratories, Liverpool University Hospitals Foundation Trusts, Liverpool, UK
| | - Karen Aughton
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Sarah E. Coupland
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Liverpool Clinical Laboratories, Liverpool University Hospitals Foundation Trusts, Liverpool, UK
| |
Collapse
|
37
|
Guidelines for uveal melanoma: a critical appraisal of systematically identified guidelines using the AGREE II and AGREE-REX instrument. J Cancer Res Clin Oncol 2020; 146:1079-1088. [PMID: 32036455 PMCID: PMC7085474 DOI: 10.1007/s00432-020-03141-w] [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: 01/02/2020] [Accepted: 01/30/2020] [Indexed: 12/30/2022]
Abstract
PURPOSE Clinical practice guidelines provide recommendations for the management of diseases. In orphan conditions such as uveal melanoma (UM), guideline developers are challenged to provide practical and useful guidance even in the absence of high-quality evidence. Here, we assessed the methodological quality and identified deficiencies of international guidelines on UM as a base for future guideline development. METHODS A systematic search was carried out in guideline databases, Medline and Embase until 27th May 2019 for guidelines on UM published between 2004 and 2019. Five independent reviewers assessed the methodological quality of the identified guidelines using the instruments "Appraisal of Guidelines for Research and Evaluation II" (AGREE II) and AGREE-REX (Recommendation EXcellence). Descriptive analysis was performed and subgroup differences were explored with the Kruskal-Wallis (H) test. The relationship between the individual domains and items of the instruments were examined using Spearman's correlation. RESULTS Five guidelines published from 2014 to 2018 by consortia of the United States of America, Canada and the United Kingdom (UK) were included. The highest scores were obtained by the UK guideline fulfilling 48-86% of criteria in AGREE II and 30-60% for AGREE-REX. All guidelines showed deficiencies in the domains "editorial independence", "applicability", and "recommendation". Subgroup differences were identified only for the domain "editorial independence". CONCLUSION The UK guideline achieved the highest scores with both instruments and may serve as a basis for future guideline development in UM. The domains "editorial independence", "recommendation", and "applicability" were identified as methodological weaknesses and require particular attention and improvement in future guidelines.
Collapse
|
38
|
Wessely A, Steeb T, Erdmann M, Heinzerling L, Vera J, Schlaak M, Berking C, Heppt MV. The Role of Immune Checkpoint Blockade in Uveal Melanoma. Int J Mol Sci 2020; 21:ijms21030879. [PMID: 32013269 PMCID: PMC7037664 DOI: 10.3390/ijms21030879] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/27/2020] [Accepted: 01/27/2020] [Indexed: 12/25/2022] Open
Abstract
Uveal melanoma (UM) represents the most common intraocular malignancy in adults and accounts for about 5% of all melanomas. Primary disease can be effectively controlled by several local therapy options, but UM has a high potential for metastatic spread, especially to the liver. Despite its clinical and genetic heterogeneity, therapy of metastatic UM has largely been adopted from cutaneous melanoma (CM) with discouraging results until now. The introduction of antibodies targeting CTLA-4 and PD-1 for immune checkpoint blockade (ICB) has revolutionized the field of cancer therapy and has achieved pioneering results in metastatic CM. Thus, expectations were high that patients with metastatic UM would also benefit from these new therapy options. This review provides a comprehensive and up-to-date overview on the role of ICB in UM. We give a summary of UM biology, its clinical features, and how it differs from CM. The results of several studies that have been investigating ICB in metastatic UM are presented. We discuss possible reasons for the lack of efficacy of ICB in UM compared to CM, highlight the pitfalls of ICB in this cancer entity, and explain why other immune-modulating therapies could still be an option for future UM therapies.
Collapse
Affiliation(s)
- Anja Wessely
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University, Ulmenweg 18, 91054 Erlangen, Germany; (A.W.); (T.S.); (M.E.); (L.H.); (J.V.); (C.B.)
| | - Theresa Steeb
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University, Ulmenweg 18, 91054 Erlangen, Germany; (A.W.); (T.S.); (M.E.); (L.H.); (J.V.); (C.B.)
| | - Michael Erdmann
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University, Ulmenweg 18, 91054 Erlangen, Germany; (A.W.); (T.S.); (M.E.); (L.H.); (J.V.); (C.B.)
| | - Lucie Heinzerling
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University, Ulmenweg 18, 91054 Erlangen, Germany; (A.W.); (T.S.); (M.E.); (L.H.); (J.V.); (C.B.)
| | - Julio Vera
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University, Ulmenweg 18, 91054 Erlangen, Germany; (A.W.); (T.S.); (M.E.); (L.H.); (J.V.); (C.B.)
| | - Max Schlaak
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Frauenlobstr. 9-11, 80337 Munich, Germany;
| | - Carola Berking
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University, Ulmenweg 18, 91054 Erlangen, Germany; (A.W.); (T.S.); (M.E.); (L.H.); (J.V.); (C.B.)
| | - Markus Vincent Heppt
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University, Ulmenweg 18, 91054 Erlangen, Germany; (A.W.); (T.S.); (M.E.); (L.H.); (J.V.); (C.B.)
- Correspondence: ; Tel.: +49-9131-85-35747
| |
Collapse
|
39
|
Seedor RS, Eschelman DJ, Gonsalves CF, Adamo RD, Orloff M, Amjad A, Sharpe-Mills E, Chervoneva I, Shields CL, Shields JA, Mastrangelo MJ, Sato T. An Outcome Assessment of a Single Institution's Longitudinal Experience with Uveal Melanoma Patients with Liver Metastasis. Cancers (Basel) 2020; 12:cancers12010117. [PMID: 31906411 PMCID: PMC7016993 DOI: 10.3390/cancers12010117] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/22/2019] [Accepted: 12/30/2019] [Indexed: 12/17/2022] Open
Abstract
There is no FDA-approved treatment for metastatic uveal melanoma (UM) and overall outcomes are generally poor for those who develop liver metastasis. We performed a retrospective single-institution chart review on consecutive series of UM patients with liver metastasis who were treated at Thomas Jefferson University Hospital between 1971–1993 (Cohort 1, n = 80), 1998–2007 (Cohort 2, n = 198), and 2008–2017 (Cohort 3, n = 452). In total, 70% of patients in Cohort 1 received only systemic therapies as their treatment modality for liver metastasis, while 98% of patients in Cohort 2 and Cohort 3 received liver-directed treatment either alone or with systemic therapy. Median Mets-to-Death OS was shortest in Cohort 1 (5.3 months, 95% CI: 4.2–7.0), longer in Cohort 2 (13.6 months, 95% CI: 12.2–16.6) and longest in Cohort 3 (17.8 months, 95% CI: 16.6–19.4). Median Eye Tx-to-Death OS was shortest in Cohort 1 (40.8 months, 95% CI: 37.1–56.9), and similar in Cohort 2 (62.6 months, 95% CI: 54.6–71.5) and Cohort 3 (59.4 months, 95% CI: 56.2–64.7). It is speculated that this could be due to the shift of treatment modalities from DTIC-based chemotherapy to liver-directed therapies. Combination of liver-directed and newly developed systemic treatments may further improve the survival of these patients.
Collapse
Affiliation(s)
- Rino S. Seedor
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - David J. Eschelman
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Carin F. Gonsalves
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Robert D. Adamo
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Marlana Orloff
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Anjum Amjad
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Erin Sharpe-Mills
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Inna Chervoneva
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Carol L. Shields
- Oncology Service, Wills Eye Hospital, Philadelphia, PA 19107, USA
| | - Jerry A. Shields
- Oncology Service, Wills Eye Hospital, Philadelphia, PA 19107, USA
| | - Michael J. Mastrangelo
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Takami Sato
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Correspondence: ; Tel.: +1-215-955-1195; Fax: +1-215-923-0797
| |
Collapse
|
40
|
Zhang J, Sun M, Hao M, Diao K, Wang J, Li S, Cao Q, Mi X. FAM53A Affects Breast Cancer Cell Proliferation, Migration, and Invasion in a p53-Dependent Manner. Front Oncol 2019; 9:1244. [PMID: 31799197 PMCID: PMC6874147 DOI: 10.3389/fonc.2019.01244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022] Open
Abstract
Family with sequence similarity 53-member A (FAM53A) is an uncharacterized protein with a suspected but unclear role in tumorigenesis. In this study, we examined its role in breast cancer. Immunohistochemical staining of specimens from 199 cases of breast cancer demonstrated that FAM53A levels were negatively correlated with p53 status. In the p53 wild-type breast cancer cell line MCF-7, FAM53A overexpression inhibited cell migration, invasion, and proliferation, downregulated the expression of Snail, cyclin D1, RhoA, RhoC, and MMP9, and decreased mitogen-activated protein kinase kinase (MEK) and extracellular-signal regulated kinase (ERK) phosphorylation. Concurrently, it upregulated E-cadherin and p21 expression levels. Interestingly, opposite trends were observed in the p53-null breast cancer cell line MDA-MB-231. The MEK inhibitor PD98059 reduced the biological effects of FAM53A knockdown in MCF-7 cells and FAM53A overexpression in MDA-MB-231 cells, suggesting that FAM53A affects breast cancer through the MEK-ERK pathway. Silencing TP53 in MCF-7 cells and stably expressing wild-type p53 in MDA-MB-231 cells confirmed that the effects of FAM53A signaling through the MEK/ERK pathway depended on the p53 status of the cells. These results suggest that FAM53A acts as a tumor suppressor in p53-positive breast cancer by modulating the MEK-ERK pathway, but may be a potential candidate for targeted anticancer therapies in p53-negative breast cancer.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Mingfang Sun
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Miaomiao Hao
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Kexin Diao
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Jian Wang
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Shiping Li
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Qixue Cao
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaoyi Mi
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
41
|
Heppt MV, Amaral T, Kähler KC, Heinzerling L, Hassel JC, Meissner M, Kreuzberg N, Loquai C, Reinhardt L, Utikal J, Dabrowski E, Gesierich A, Pföhler C, Terheyden P, Thoms KM, Zimmer L, Eigentler TK, Kirchberger MC, Stege HM, Meier F, Schlaak M, Berking C. Combined immune checkpoint blockade for metastatic uveal melanoma: a retrospective, multi-center study. J Immunother Cancer 2019; 7:299. [PMID: 31722735 PMCID: PMC6854774 DOI: 10.1186/s40425-019-0800-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 10/30/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Uveal melanoma (UM) is highly refractory to treatment with dismal prognosis in advanced stages. The value of the combined checkpoint blockade with CTLA-4 and PD-1 inhibition in metastatic UM is currently unclear. METHODS Patients with metastatic or unresectable UM treated with ipilimumab in combination with a PD-1 inhibitor were collected from 16 German skin cancer centers. Patient records of 64 cases were analyzed for response, progression-free survival (PFS), overall survival (OS), and safety. Clinical parameters and serum biomarkers associated with OS and treatment response were determined with Cox regression modelling and logistic regression. RESULTS The best overall response rate to combined checkpoint blockade was 15.6% with 3.1 and 12.5% complete and partial response, respectively. The median duration of response was 25.5 months (range 9.0-65.0). Stable disease was achieved in 21.9%, resulting in a disease control rate of 37.5% with a median duration of the clinical benefit of 28.0 months (range 7.0-65.0). The median PFS was 3.0 months (95% CI 2.4-3.6). The median OS was estimated to 16.1 months (95% CI 12.9-19.3). Regarding safety, 39.1% of treated patients experienced a severe, treatment-related adverse event according to the CTCAE criteria (grade 3: 37.5%; grade 4: 1.6%). The most common toxicities were colitis (20.3%), hepatitis (20.3%), thyreoiditis (15.6%), and hypophysitis (7.8%). A poor ECOG performance status was an independent risk factor for decreased OS (p = 0.007). CONCLUSIONS The tolerability of the combined checkpoint blockade in UM may possibly be better than in trials on cutaneous melanoma. This study implies that combined checkpoint blockade represents the hitherto most effective treatment option available for metastatic UM available outside of clinical trials.
Collapse
Affiliation(s)
- Markus V Heppt
- Department of Dermatology and Allergy, Munich University Hospital (LMU), Frauenlobstr. 9-11, 80337, Munich, Germany
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany
| | - Teresa Amaral
- Department of Dermatology, Center for Dermatooncology, University Hospital Tübingen, Liebermeisterstr. 25, 72076, Tübingen, Germany
- Portuguese Air Force Health Care Direction, Lisbon, Portugal
| | - Katharina C Kähler
- Department of Dermatology, University Hospital Schleswig-Holstein, Campus Kiel, Rosalind-Franklin-Str. 7, 24105, Kiel, Germany
| | - Lucie Heinzerling
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany
| | - Jessica C Hassel
- Skin Cancer Center, Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Markus Meissner
- Department of Dermatology, Venereology and Allergology, Goethe University, Theodor-Stern Kai 7, 60590, Frankfurt am Main, Germany
| | - Nicole Kreuzberg
- Department of Dermatology and Venereology, Skin Cancer Center at the Center of Integrated Oncology (CIO) Köln Bonn, University Hospital of Cologne, Kerpenerstr. 62, 50937, Cologne, Germany
| | - Carmen Loquai
- Department of Dermatology, University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Lydia Reinhardt
- Department of Dermatology, Skin Cancer Center, Medical Faculty and University Hospital Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Evelyn Dabrowski
- Department of Dermatology, Klinikum Ludwigshafen, Bremserstr. 79, 67063, Ludwigshafen, Germany
| | - Anja Gesierich
- Department of Dermatology, University Hospital Würzburg, Josef-Schneider Straße 2, 97080, Würzburg, Germany
| | - Claudia Pföhler
- Department of Dermatology, Saarland University Medical School, Kirrbergerstr, 66421, Homburg/Saar, Germany
| | - Patrick Terheyden
- Department of Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Kai-Martin Thoms
- Department of Dermatology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital, University Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Thomas K Eigentler
- Department of Dermatology, Center for Dermatooncology, University Hospital Tübingen, Liebermeisterstr. 25, 72076, Tübingen, Germany
| | - Michael C Kirchberger
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany
| | - Henner M Stege
- Department of Dermatology, University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Friedegund Meier
- Department of Dermatology, Skin Cancer Center, Medical Faculty and University Hospital Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Max Schlaak
- Department of Dermatology and Allergy, Munich University Hospital (LMU), Frauenlobstr. 9-11, 80337, Munich, Germany
| | - Carola Berking
- Department of Dermatology and Allergy, Munich University Hospital (LMU), Frauenlobstr. 9-11, 80337, Munich, Germany.
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany.
| |
Collapse
|
42
|
Increased Non-Homologous End Joining Makes DNA-PK a Promising Target for Therapeutic Intervention in Uveal Melanoma. Cancers (Basel) 2019; 11:cancers11091278. [PMID: 31480356 PMCID: PMC6769470 DOI: 10.3390/cancers11091278] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 02/07/2023] Open
Abstract
Uveal melanoma (UM) is the most common primary intraocular tumour in adults, with a mean survival of six months following metastasis. The survival rates have not improved in over 30 years. This study has shown that sister chromatid exchange (SCE) is low in UM which is likely due to a reduced expression of FANCD2. As FANCD2 can function to suppress non-homologous end joining (NHEJ), this study therefore investigated NHEJ in UM. The activation of the catalytic subunit of the NHEJ pathway protein DNA-dependent protein kinase (DNA-PK) was measured by analysing the foci formation and the ligation efficiency by NHEJ determined using a plasmid-based end-joining assay. Using small-interfering RNA (siRNA) knock-down, and chemical inhibitors of DNA-PK, the survival of primary UM cultures and two cell lines were determined. To assess the homologous recombination capacity in response to the inhibition of DNA-PK, a SCE analysis was performed. In addition, to support the findings, the messenger RNA (mRNA) expression of genes associated with NHEJ was analysed using the Cancer Genome Atlas (TCGA)-UM RNAseq data (n = 79). The NHEJ activity and DNA-PKcs activation was upregulated in UM and the inhibition of DNA-PK selectively induced apoptosis and sensitized to ionising radiation and inter-strand cross-linking agents. The inhibition of the NHEJ protein DNA-PK is lethal to UM, indicating a potentially effective therapeutic option, either alone or as a sensitizer for other treatments.
Collapse
|
43
|
Musi E, Schwartz GK, Yoo JH, Odelberg SJ, Li DY, Bonner MY, Selvakumar P, Rao S, Gilbert LC, Elsey J, Arbiser JL. Tris DBA palladium is an orally available inhibitor of GNAQ mutant uveal melanoma in vivo. Oncotarget 2019; 10:4424-4436. [PMID: 31320995 PMCID: PMC6633893 DOI: 10.18632/oncotarget.27040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/05/2019] [Indexed: 12/22/2022] Open
Abstract
Uveal melanoma is a rare but often lethal malignancy and is the leading cause of death due to an ophthalmic condition. Uveal melanoma is often diagnosed at a late stage and has a strong propensity to hepatic metastasis. Recently, the most common driver mutations in uveal melanoma have been identified, predominantly in the G-proteins GNAQ. This pattern differs from that of cutaneous melanoma in which Braf and Nras predominate. There are no current clinically used agents that target GNAQ mutations, unlike the use of Braf inhibitors in cutaneous melanoma. We tested the novel agent Tris DBA palladium and found that it was markedly more effective against GNAQ mutant melanomas than wild type uveal melanomas. Given that ARF6 has recently been discovered as a node in GNAQ mutations, we evaluated the efficacy of Tris DBA palladium on ARF6 signaling and found that it was effective in inhibiting ARF6 activation. Finally, Tris DBA palladium was orally effective against GNAQ mutant melanoma in vivo. Tris DBA Palladium deserves further evaluation as a systemic agent for uveal melanoma.
Collapse
Affiliation(s)
- Elgilda Musi
- Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Gary K. Schwartz
- Department of Medicine, Columbia University Medical Center, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University College of Medicine, New York, New York, USA
| | - Jae Hyuk Yoo
- Department of Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Shannon J. Odelberg
- Department of Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Dean Y. Li
- Department of Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Michael Y. Bonner
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ponniah Selvakumar
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Shikha Rao
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Linda C. Gilbert
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia, USA
- Veterans Affairs Medical Center, Decatur, Georgia, USA
| | - Justin Elsey
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jack L. Arbiser
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia, USA
- Veterans Affairs Medical Center, Decatur, Georgia, USA
| |
Collapse
|
44
|
Croce M, Ferrini S, Pfeffer U, Gangemi R. Targeted Therapy of Uveal Melanoma: Recent Failures and New Perspectives. Cancers (Basel) 2019; 11:E846. [PMID: 31216772 PMCID: PMC6628160 DOI: 10.3390/cancers11060846] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/23/2022] Open
Abstract
Among Uveal Melanoma (UM) driver mutations, those involving GNAQ or GNA11 genes are the most frequent, while a minor fraction of tumors bears mutations in the PLCB4 or CYSLTR2 genes. Direct inhibition of constitutively active oncoproteins deriving from these mutations is still in its infancy in UM, whereas BRAFV600E-targeted therapy has obtained relevant results in cutaneous melanoma. However, UM driver mutations converge on common downstream signaling pathways such as PKC/MAPK, PI3K/AKT, and YAP/TAZ, which are presently considered as actionable targets. In addition, BAP1 loss, which characterizes UM metastatic progression, affects chromatin structure via histone H2A deubiquitylation that may be counteracted by histone deacetylase inhibitors. Encouraging results of preclinical studies targeting signaling molecules such as MAPK and PKC were unfortunately not confirmed in early clinical studies. Indeed, a general survey of all clinical trials applying new targeted and immune therapy to UM displayed disappointing results. This paper summarizes the most recent studies of UM-targeted therapies, analyzing the possible origins of failures. We also focus on hyperexpressed molecules involved in UM aggressiveness as potential new targets for therapy.
Collapse
Affiliation(s)
- Michela Croce
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| | | | - Ulrich Pfeffer
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| | | |
Collapse
|