1
|
Wu DH, Lyu CG, Zhao D, Yang CG, Liu SQ, Zhu JT, Yang YL, Guo LP, Kang CZ. Ardisia Crispae Radix et Rhizoma: A review of botany, traditional uses, phytochemistry, pharmacology, and toxicology. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119093. [PMID: 39557103 DOI: 10.1016/j.jep.2024.119093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/09/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ardisia Crispae Radix et Rhizoma comprises three primary source plants: Ardisia crenata Sims, Ardisia crispa (Thunb.) A. DC., and Ardisia crenata Sims var. bicolor (Walk) C. Y. Wu et C. Chen. These species are prevalent in mid-to-low-latitude regions and are traditionally utilized as herbal medicines in East Asia and India. They have demonstrated notable efficacy in anti-inflammatory properties and possess potential anti-tumor, antimicrobial, antioxidant, and anti-angiogenic activities. AIM This review systematically evaluated the botany, traditional uses, phytochemistry, pharmacology, and toxicity of Ardisia Crispae Radix et Rhizoma to assist future innovative research and facilitate the development of new therapeutic agents. MATERIALS AND METHODS Literature was sourced from both library and electronic databases, including Web of Science, PubMed, Elsevier, Google Scholar, and CNKI. The review focuses on phytochemistry, pharmacological research, and toxicity studies related to Ardisia Crispae Radix et Rhizoma. RESULTS Ardisia Crispae Radix et Rhizoma is traditionally used to treat inflammation and injuries. Current pharmacological studies suggest its potential anti-tumor, anti-inflammatory, antimicrobial, antioxidant, and anti-angiogenic activities. This review identified approximately 161 compounds in Ardisia Crispae Radix et Rhizoma, such as saponins, flavonoids, coumarins, essential oils, lignans, and terpenes. CONCLUSIONS Ardisia Crispae Radix et Rhizoma is a promising medicinal plant resource with a diverse range of specialized metabolites. However, existing research predominately addresses phytochemistry and pharmacological activities, with limited exploration of underlying mechanisms. Further systematic evaluations of its efficacy and clinical safety are required.
Collapse
Affiliation(s)
- De-Hua Wu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China; Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Chao-Geng Lyu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Dan Zhao
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Chang-Gui Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Si-Qi Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ji-Tong Zhu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ya-Ling Yang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Lan-Ping Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Chuan-Zhi Kang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
2
|
Khera E, Dharmarajan L, Hainzl D, Engelhardt V, Vostiarova H, Davis J, Ebel N, Wuersch K, Romanet V, Sharaby S, Kearns JD. QSP modeling of a transiently inactivating antibody-drug conjugate highlights benefit of short antibody half life. J Pharmacokinet Pharmacodyn 2024; 52:7. [PMID: 39690276 DOI: 10.1007/s10928-024-09956-1] [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: 05/14/2024] [Accepted: 10/03/2024] [Indexed: 12/19/2024]
Abstract
Antibody drug conjugates (ADC) are a promising class of oncology therapeutics consisting of an antibody conjugated to a payload via a linker. DYP688 is a novel ADC comprising of a signaling protein inhibitor payload (FR900359) that undergoes unique on-antibody inactivation in plasma, resulting in complex pharmacology. To assess the impact of FR inactivation on DYP688 pharmacology and clinical developability, we performed translational modeling of preclinical PK and tumor growth inhibition (TGI) data, accompanied by mechanistic Krogh cylinder tumor modeling. Using a PK-TGI model, we identified a composite exposure-above-tumorostatic concentration (AUCTSC) metric as the PK-driver of efficacy. To underpin the mechanisms behind AUCTSC as the driver of efficacy, we performed quantitative systems pharmacology (QSP) modeling of DYP688 intratumoral pharmacokinetics and pharmacodynamics. Through exploratory simulations, we show that by deviating from canonical ADC design dogma, DYP688 has optimal FR900359 activity despite its transient inactivation. Finally, we performed the successful preclinical to clinical translation of DYP688 PK, including the payload inactivation kinetics, evidenced by good agreement of the predicted PK to the observed interim clinical PK. Overall, this work highlights early quantitative pharmacokinetics as a missing link in the ADC design-developability chasm.
Collapse
Affiliation(s)
- Eshita Khera
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Cambridge, MA, USA
| | - Lekshmi Dharmarajan
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Basel, Switzerland
| | - Dominik Hainzl
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Cambridge, MA, USA
| | - Volker Engelhardt
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Basel, Switzerland
| | - Helena Vostiarova
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Basel, Switzerland
| | - John Davis
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Cambridge, MA, USA
| | - Nicolas Ebel
- Oncology, Novartis Biomedical Research, Basel, Switzerland
| | - Kuno Wuersch
- Preclinical Safety, Novartis Biomedical Research, Basel, Switzerland
| | | | - Sherif Sharaby
- PK Sciences, Translational Medicine, Novartis Biomedical Research, East Hanover, NJ, USA
| | - Jeffrey D Kearns
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Cambridge, MA, USA.
| |
Collapse
|
3
|
Ouyang S, Shi S, Ding W, Ge Y, Su Y, Mo J, Peng K, Zhang Q, Liu G, Xiao W, Yue P, Lu J, Wang Y, Xiong X, Zhang X. Neuropeptide Precursor VGF Promotes Liver Metastatic Colonization of Gαq Mutant Uveal Melanoma by Facilitating Tumor Microenvironment via Paracrine Loops. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407967. [PMID: 39422674 PMCID: PMC11633529 DOI: 10.1002/advs.202407967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Uveal melanoma (UM), the predominant primary ocular malignancy, often progresses to liver metastasis with limited therapeutic options. The interplay of the tumor microenvironment, encompassing secreted soluble factors, plays a crucial role in facilitating liver metastasis. In this study, the role is elucidated of the neural growth factor-inducible gene (VGF), a secreted neuropeptide precursor, in Gαq mutant UM. Employing a multiomics approach, encompassing transcriptomic and secretomic analyses, the intricate involvement of VGF in UM progression is unveiled. VGF is upregulated in Gαq mutant UM cells and associated with poor prognosis of UM patients. Targeting VGF significantly suppressed the growth of UM in vitro and in vivo. Further evidence shows that VGF is regulated by Gαq through MAPK/CREB pathway. Mechanistically, CREB modulates VGF expression by directly binding to consensus DNA response elements in the promoters of the VGF gene. Combined inhibition of Gαq and MEK remarkably reduces tumor burden in the UM xenograft model. Notably, VGF triggers liver metastatic colonization of UM and activates the fibrosis of hepatic stellate cells (HSCs), creating a favorable microenvironment, through an autocrine and paracrine loop. Furthermore, VGF directly binds to TGFBR2 and regulates TGF-β-SMAD signaling pathway, thereby regulating genes associated with endothelial-mesenchymal transition (EMT) to promote metastasis. Taken together, these findings identify VGF as a pivotal driver in the progression and metastasis of Gαq mutant UM and confers a promising therapeutic target and strategy for UM patients.
Collapse
Affiliation(s)
- Shumin Ouyang
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Shuo Shi
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Wen Ding
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Yang Ge
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Yingxue Su
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhou510060China
| | - Jianshan Mo
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Keren Peng
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Qiyi Zhang
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Guopin Liu
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhou510060China
| | - Wei Xiao
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhou510060China
| | - Peibin Yue
- Department of MedicineDivision of Hematology‐Oncologyand Samuel Oschin Comprehensive Cancer InstituteCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Jinjian Lu
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauMacao999078China
| | - Yandong Wang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhou510060China
| | - Xiaofeng Xiong
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Xiaolei Zhang
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| |
Collapse
|
4
|
Dong J, Xu Y, Yu D, Zhang X, Wang A, Lv L, Li Z. Gq/G11 oncogenic mutations promote PD-L1 expression and suppress tumor immunity. Eur J Cell Biol 2024; 103:151467. [PMID: 39550833 DOI: 10.1016/j.ejcb.2024.151467] [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: 05/20/2024] [Revised: 10/24/2024] [Accepted: 11/08/2024] [Indexed: 11/19/2024] Open
Abstract
Uveal melanoma (UM) is the predominant form of eye cancer. The genes GNAQ and GNA11, encoding Gq and G11 respectively, are most frequently mutated in UM and are considered the major drivers of UM carcinogenesis by activating YAP. However, the mechanisms by which metastatic UM evades the immune system remain poorly understood. In this study, we found that oncogenic mutations of Gq/G11 promoted YAP and PD-L1 expression, modifying the tumor microenvironment and promoting immune evasion of UM. Consistently, the levels of GNAQ/GNA11 and YAP positively correlated to PD-L1 expression in UM patients. Furthermore, silencing YAP or treating with its inhibitor, Verteporfin, attenuated PD-L1 expression induced by Gq/G11 mutations, thereby enhancing T cell activation and T cell-mediated cytotoxicity. Collectively, this study reveals a potential role of Gq/G11 mutations on immune evasion of UM, a new mechanism of Gq/11 mutations-induced tumorigenesis, highlighting Gq/G11 and YAP as potential immunotherapeutic targets and suggesting Verteporfin as an adjuvant for immunotherapy of UM patients with GNAQ or GNA11 mutations.
Collapse
Affiliation(s)
- Jingyan Dong
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China; Shanxi Eye Hospital, Taiyuan 030001, China
| | - Yue Xu
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200032, China
| | - Dawei Yu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China
| | - Xiaoling Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China
| | - Anqi Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China
| | - Lei Lv
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Zhiqing Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China.
| |
Collapse
|
5
|
Pistorius D, Richard E, Buntin K, Dresen K, Wollbrett S, Weber E, Haberkorn A, Manchado E, Petersen F. Condensation Domain Editing of the FR900359 Assembly Line Yields a Novel Analog Amenable to Late-Stage Functionalization. Chembiochem 2024; 25:e202400491. [PMID: 39076125 DOI: 10.1002/cbic.202400491] [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: 06/05/2024] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 07/31/2024]
Abstract
The natural product FR900359 (FR) has generated significant attention lately, due to its characteristics as potent and selective inhibitor of Gq/11 mediated signal transduction of associated G protein-coupled receptors (GPCRs). This makes FR both a widely used pharmacological tool compound and a lead molecule for targeted cancer therapy. The exploration of structure-activity-relationship (SAR) of the scaffold by total synthesis has been complicated by its structural complexity and its incompatibility with standard approaches of solid-phase peptide synthesis. Options for late-stage functionalization of FR are limited due to a lack of tractable functional groups. Here we present a mixed approach combining (i) genetic engineering of the FR-assembly line in Chromobacterium vaccinii, to obtain a novel FR analog featuring a primary amine, with (ii) its subsequent synthetic modification and biological profiling for further SAR exploration of the FR scaffold.
Collapse
Affiliation(s)
- Dominik Pistorius
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Etienne Richard
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Kathrin Buntin
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Kathrin Dresen
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Séverine Wollbrett
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Eric Weber
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Anne Haberkorn
- Oncology Disease Area, Biomedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Eusebio Manchado
- Oncology Disease Area, Biomedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Frank Petersen
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| |
Collapse
|
6
|
Dwyer MB, Aumiller JL, Wedegaertner PB. Going Rogue: Mechanisms, Regulation, and Roles of Mutationally Activated G α in Human Cancer. Mol Pharmacol 2024; 106:198-215. [PMID: 39187387 PMCID: PMC11493338 DOI: 10.1124/molpharm.124.000743] [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] [Received: 08/01/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024] Open
Abstract
G protein-coupled receptors (GPCRs) couple to heterotrimeric G proteins, comprised of α and βγ subunits, to convert extracellular signals into activation of intracellular signaling pathways. Canonically, GPCR-mediated activation results in the exchange of GDP for GTP on G protein α subunits (Gα) and the dissociation of Gα-GTP and G protein βγ subunits (Gβγ), both of which can regulate a variety of signaling pathways. Hydrolysis of bound GTP by Gα returns the protein to Gα-GDP and allows reassociation with Gβγ to reform the inactive heterotrimer. Naturally occurring mutations in Gα have been found at conserved glutamine and arginine amino acids that disrupt the canonical G protein cycle by inhibiting GTP hydrolysis, rendering these mutants constitutively active. Interestingly, these dysregulated Gα mutants are found in many different cancers due to their ability to sustain aberrant signaling without a need for activation by GPCRs. This review will highlight an increased recognition of the prevalence of such constitutively activating Gα mutations in cancers and the signaling pathways activated. In addition, we will discuss new knowledge regarding how these constitutively active Gα are regulated, how different mutations are biochemically distinct, and how mutationally activated Gα are unique compared with GPCR-activated Gα Lastly, we will discuss recent progress in developing inhibitors directly targeting constitutively active Gα mutants. SIGNIFICANCE STATEMENT: Constitutively activating mutations in G protein α subunits (Gα) widely occur in and contribute to the development of many human cancers. To develop ways to inhibit dysregulated, oncogenic signaling by these mutant Gα, it is crucial to better understand mechanisms that lead to constitutive Gα activation and unique mechanisms that regulate mutationally activated Gα in cells. The prevalence of activating mutations in Gα in various cancers makes Gα proteins compelling targets for the development of therapeutics.
Collapse
Affiliation(s)
- Morgan B Dwyer
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jenna L Aumiller
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Philip B Wedegaertner
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
7
|
Yenisehirli G, Borges S, Braun SS, Zuniga AN, Quintana GI, Kutsnetsoff JN, Rodriguez S, Adis EV, Lopez S, Dollar JJ, Stathias V, Volmar CH, Karaca E, Brothers S, Bilbao D, Harbour JW, Correa ZM, Kurtenbach S. Identification of targetable epigenetic vulnerabilities in uveal melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617464. [PMID: 39416076 PMCID: PMC11482939 DOI: 10.1101/2024.10.11.617464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Uveal melanoma (UM) is the most prevalent primary intraocular malignancy in adults, which preferentially metastasizes to the liver in approximately half of all cases. Metastatic UM is notoriously resistant to therapy and is almost uniformly fatal. UM metastasis is most strongly associated with mutational inactivation of the BAP1 tumor suppressor gene. Given the role of BAP1 in epigenetic regulation as the ubiquitin hydrolase subunit of the polycomb repressive deubiquitinase (PR-DUB) complex, we conducted high-throughput drug screening using a well-characterized epigenetic compound library to identify new therapeutic vulnerabilities. We identified several promising new lead compounds, in particular the extra-terminal domain protein (BET) inhibitor mivebresib (ABBV-075). Mivebresib significantly improved survival rates in a metastatic uveal melanoma xenograft mouse model and entirely prevented detectable metastases to the bones, spinal cord, and brain. RNA sequencing revealed a notable overlap between the genes and pathways affected by HDAC and BET inhibition, including the reversal of gene signatures linked to high metastatic risk and upregulation of genes associated with a neuronal phenotype. Together, we found that UM cells are particularly vulnerable to class I HDAC and BET inhibition, and highlight the BET inhibitor mivebresib as a promising candidate for further clinical evaluation.
Collapse
Affiliation(s)
- Gulum Yenisehirli
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Sebastian Borges
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Steffanie S. Braun
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Ashley N. Zuniga
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Gabriela I. Quintana
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Jeffim N. Kutsnetsoff
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Sara Rodriguez
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Emily V. Adis
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Sofia Lopez
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - James J. Dollar
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Vasileios Stathias
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
| | - Claude H. Volmar
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine
| | - Efe Karaca
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine
| | - Shaun Brothers
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine
| | - Daniel Bilbao
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine
| | - J. William Harbour
- Department of Ophthalmology and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center
| | - Zelia M. Correa
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| | - Stefan Kurtenbach
- Department of Ophthalmology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
- Interdisciplinary Stem Cell Institute (ISCI), University of Miami Miller School of Medicine
| |
Collapse
|
8
|
Huber T, Horioka-Duplix M, Chen Y, Saca VR, Ceraudo E, Chen Y, Sakmar TP. The role of signaling pathways mediated by the GPCRs CysLTR1/2 in melanocyte proliferation and senescence. Sci Signal 2024; 17:eadp3967. [PMID: 39288219 DOI: 10.1126/scisignal.adp3967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024]
Abstract
In contrast with sun exposure-induced melanoma, rarer melanocytic tumors and neoplasms with low mutational burden present opportunities to study isolated signaling mechanisms. These include uveal melanoma and blue nevi, which are often driven by mutations within the G protein-coupled signaling cascade downstream of cysteinyl leukotriene receptor 2. Here, we review how the same mutations within this pathway drive the growth of melanocytes in one tissue but can inhibit the growth of those in another, exemplifying the role of the tissue environment in the delicate balance between uncontrolled cell growth and senescence.
Collapse
Affiliation(s)
- Thomas Huber
- Laboratory of Chemical Biology and Signal Transduction, Rockefeller University, New York, NY 10065, USA
| | - Mizuho Horioka-Duplix
- Laboratory of Chemical Biology and Signal Transduction, Rockefeller University, New York, NY 10065, USA
- Tri-Institutional PhD Program in Chemical Biology, New York, NY 10065, USA
| | - Yuanhuang Chen
- Laboratory of Chemical Biology and Signal Transduction, Rockefeller University, New York, NY 10065, USA
- Tri-Institutional PhD Program in Chemical Biology, New York, NY 10065, USA
| | - Victoria R Saca
- Laboratory of Chemical Biology and Signal Transduction, Rockefeller University, New York, NY 10065, USA
- Tri-Institutional PhD Program in Chemical Biology, New York, NY 10065, USA
| | - Emilie Ceraudo
- Laboratory of Chemical Biology and Signal Transduction, Rockefeller University, New York, NY 10065, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
9
|
Bonifer C, Hanke W, Mühle J, Löhr F, Becker-Baldus J, Nagel J, Schertler GFX, Müller CE, König GM, Hilger D, Glaubitz C. Structural response of G protein binding to the cyclodepsipeptide inhibitor FR900359 probed by NMR spectroscopy. Chem Sci 2024; 15:12939-12956. [PMID: 39148790 PMCID: PMC11323312 DOI: 10.1039/d4sc01950d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/27/2024] [Indexed: 08/17/2024] Open
Abstract
The cyclodepsipeptide FR900359 (FR) and its analogs are able to selectively inhibit the class of Gq proteins by blocking GDP/GTP exchange. The inhibitor binding site of Gq has been characterized by X-ray crystallography, and various binding and functional studies have determined binding kinetics and mode of inhibition. Here we investigate isotope-labeled FR bound to the membrane-anchored G protein heterotrimer by solid-state nuclear magnetic resonance (ssNMR) and in solution by liquid-state NMR. The resulting data allowed us to identify regions of the inhibitor which show especially pronounced effects upon binding and revealed a generally rigid binding mode in the cis conformation under native-like conditions. The inclusion of the membrane environment allowed us to show a deep penetration of FR into the lipid bilayer illustrating a possible access mode of FR into the cell. Dynamic nuclear polarization (DNP)-enhanced ssNMR was used to observe the structural response of specific segments of the Gα subunit to inhibitor binding. This revealed rigidification of the switch I binding site and an allosteric response in the α5 helix as well as suppression of structural changes induced by nucleotide exchange due to inhibition by FR. Our NMR studies of the FR-G protein complex conducted directly within a native membrane environment provide important insights into the inhibitors access via the lipid membrane, binding mode, and structural allosteric effects.
Collapse
Affiliation(s)
- Christian Bonifer
- Institute of Biophysical Chemistry, Centre of Biomolecular Magnetic Resonance, Goethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt Germany
| | - Wiebke Hanke
- Institute for Pharmaceutical Biology, University of Bonn Nussallee 6 53115 Bonn Germany
| | - Jonas Mühle
- Division of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institute Forschungsstr. 111, 5232 Villigen PSI Switzerland
| | - Frank Löhr
- Institute of Biophysical Chemistry, Centre of Biomolecular Magnetic Resonance, Goethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt Germany
| | - Johanna Becker-Baldus
- Institute of Biophysical Chemistry, Centre of Biomolecular Magnetic Resonance, Goethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt Germany
| | - Jessica Nagel
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn An der Immenburg 4 53121 Bonn Germany
| | - Gebhard F X Schertler
- Division of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institute Forschungsstr. 111, 5232 Villigen PSI Switzerland
| | - Christa E Müller
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn An der Immenburg 4 53121 Bonn Germany
| | - Gabriele M König
- Institute for Pharmaceutical Biology, University of Bonn Nussallee 6 53115 Bonn Germany
| | - Daniel Hilger
- Department of Pharmaceutical Chemistry, University of Marburg 35037 Marburg Germany
| | - Clemens Glaubitz
- Institute of Biophysical Chemistry, Centre of Biomolecular Magnetic Resonance, Goethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt Germany
| |
Collapse
|
10
|
Bustamante P, Coblentz J, Mastromonaco C, Youhnovska E, Ito H, Proença RP, Fonseca C, Dickinson K, Marcotte E, MacDonald M, Toledo-Dias AB, Bergeron S, Goyeneche A, Schmidt Andujar RA, Tsering T, Laskaris A, Jin E, Nadeau A, Porraccio T, Burnier MN, Burnier JV. Comprehensive clinical imaging, histopathological analysis and liquid biopsy-based surveillance of human uveal melanoma in a prolonged rabbit xenograft model. Melanoma Res 2024; 34:285-295. [PMID: 38847739 DOI: 10.1097/cmr.0000000000000964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Uveal melanoma is the most common intraocular tumor in adults. Our group has previously developed a human uveal melanoma animal model; however, adverse effects caused by the immunosuppressive agent, cyclosporine A, prevented animals from surviving more than 12 weeks. In this study, we tested multiple cyclosporine A doses over an extended disease course up to 20 weeks, providing complete clinical imaging of intraocular tumors, histopathological analysis and liquid biopsy biomarker analysis. Twenty albino rabbits were divided into four groups with different daily cyclosporine A schedules (0-10 mg/kg) and inoculated with human uveal melanoma cell lines, 92.1 or MP41, into the suprachoroidal space. Rabbits were monitored with fundoscopy, ultrasound and optical coherence tomography. Intraocular tumors (macroscopic or microscopic) were detected in all study animals. Tumor size and growth were correlated to cyclosporine A dose, with tumors regressing when cyclosporine A was arrested. All tumors expressed HMB-45 and MelanA; however, tumor size, pigmentation and cell morphology differed in 92.1 vs. MP41 tumors. Finally, across all groups, circulating tumor DNA from plasma and aqueous humor was detected earlier than tumor detection by imaging and correlated to tumor growth. In conclusion, using three clinically relevant imaging modalities (fundoscopy, ultrasonography and optical coherence tomography) and liquid biopsy, we were successfully able to monitor tumor progression in our rabbit xenograft model of human uveal melanoma.
Collapse
Affiliation(s)
- Prisca Bustamante
- Cancer Research Program, Research Institute of the McGill University Health Centre
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University
- Department of Pathology, McGill University, Montréal, Canada
| | - Jacqueline Coblentz
- Cancer Research Program, Research Institute of the McGill University Health Centre
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University
| | - Christina Mastromonaco
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University
| | - Emma Youhnovska
- Cancer Research Program, Research Institute of the McGill University Health Centre
| | - Hiroaki Ito
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Rita Pinto Proença
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University
- Coimbra University Hospital Center, Coimbra
- Faculty of Medicine, University of Lisbon, Lisbon
- Hospital CUF Cascais, Cascais, Portugal
| | - Cristina Fonseca
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University
- Coimbra University Hospital Center, Coimbra
| | - Kyle Dickinson
- Cancer Research Program, Research Institute of the McGill University Health Centre
| | - Emily Marcotte
- Cancer Research Program, Research Institute of the McGill University Health Centre
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University
| | - Myriam MacDonald
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University
| | | | - Sabrina Bergeron
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University
| | - Alicia Goyeneche
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University
| | | | - Thupten Tsering
- Cancer Research Program, Research Institute of the McGill University Health Centre
- Department of Pathology, McGill University, Montréal, Canada
| | - Alexander Laskaris
- Cancer Research Program, Research Institute of the McGill University Health Centre
- Department of Pathology, McGill University, Montréal, Canada
| | - Eva Jin
- Cancer Research Program, Research Institute of the McGill University Health Centre
- Department of Pathology, McGill University, Montréal, Canada
| | - Amélie Nadeau
- Cancer Research Program, Research Institute of the McGill University Health Centre
- Department of Pathology, McGill University, Montréal, Canada
| | - Tiffany Porraccio
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University
| | - Miguel N Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University
- Department of Pathology, McGill University, Montréal, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre
- Department of Pathology, McGill University, Montréal, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montréal, Canada
| |
Collapse
|
11
|
Lan J, Wu Q, Huang N, Zhang H, Yang Y, Chen L, Zhou N, He X. Identification of sulfakinin receptor regulating feeding behavior and hemolymph trehalose homeostasis in the silkworm, Bombyx mori. Sci Rep 2024; 14:14191. [PMID: 38902334 PMCID: PMC11190223 DOI: 10.1038/s41598-024-65177-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/18/2024] [Indexed: 06/22/2024] Open
Abstract
Feeding behavior, the most fundamental physiological activity, is controlled by two opposing groups of factors, orexigenic and anorexigenic factors. The sulfakinin family, an insect analogue of the mammalian satiety factor cholecystokinin (CCK), has been shown to suppress food intake in various insects. Nevertheless, the mechanisms through which sulfakinin regulates feeding behavior remain a biological question. This study aimed to elucidate the signaling pathway mediated by the anorexigenic peptide sulfakinin in Bombyx mori. We identified the Bombyx mori neuropeptide G protein-coupled receptor A9 (BNGR-A9) as the receptor for sulfakinin through functional assays. Stimulation with sulfakinin triggered a swift increase in intracellular IP3, Ca2+, and a notable enhancement of ERK1/2 phosphorylation, in a manner sensitive to a Gαq-specific inhibitor. Treatment with synthetic sulfakinin resulted in decreased food consumption and average body weight. Additionally, administering synthetic sulfakinin to silkworms significantly elevated hemolymph trehalose levels, an effect markedly reduced by pre-treatment with BNGR-A9 dsRNA. Consequently, our findings establish the sulfakinin/BNGR-A9 signaling pathway as a critical regulator of feeding behavior and hemolymph trehalose homeostasis in Bombyx mori, highlighting its roles in the negative control of food intake and the positive regulation of energy balance.
Collapse
Affiliation(s)
- Jiajing Lan
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, 311399, China
| | - Qi Wu
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212018, Jiangsu, China
| | - Nan Huang
- Department of Clinical Laboratory, The First People's Hospital of Lin'an District, Hangzhou, 311399, Zhejiang, China
| | - Hong Zhang
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, 311399, China
| | - Yuanfei Yang
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, 311399, China
| | - Linjie Chen
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, 311399, China
| | - Naiming Zhou
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Xiaobai He
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, 311399, China.
| |
Collapse
|
12
|
Fuentes-Rodriguez A, Mitchell A, Guérin SL, Landreville S. Recent Advances in Molecular and Genetic Research on Uveal Melanoma. Cells 2024; 13:1023. [PMID: 38920653 PMCID: PMC11201764 DOI: 10.3390/cells13121023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/27/2024] Open
Abstract
Uveal melanoma (UM), a distinct subtype of melanoma, presents unique challenges in its clinical management due to its complex molecular landscape and tendency for liver metastasis. This review highlights recent advancements in understanding the molecular pathogenesis, genetic alterations, and immune microenvironment of UM, with a focus on pivotal genes, such as GNAQ/11, BAP1, and CYSLTR2, and delves into the distinctive genetic and chromosomal classifications of UM, emphasizing the role of mutations and chromosomal rearrangements in disease progression and metastatic risk. Novel diagnostic biomarkers, including circulating tumor cells, DNA and extracellular vesicles, are discussed, offering potential non-invasive approaches for early detection and monitoring. It also explores emerging prognostic markers and their implications for patient stratification and personalized treatment strategies. Therapeutic approaches, including histone deacetylase inhibitors, MAPK pathway inhibitors, and emerging trends and concepts like CAR T-cell therapy, are evaluated for their efficacy in UM treatment. This review identifies challenges in UM research, such as the limited treatment options for metastatic UM and the need for improved prognostic tools, and suggests future directions, including the discovery of novel therapeutic targets, immunotherapeutic strategies, and advanced drug delivery systems. The review concludes by emphasizing the importance of continued research and innovation in addressing the unique challenges of UM to improve patient outcomes and develop more effective treatment strategies.
Collapse
Affiliation(s)
- Aurélie Fuentes-Rodriguez
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| | - Andrew Mitchell
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| | - Sylvain L. Guérin
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
| | - Solange Landreville
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| |
Collapse
|
13
|
Voss JH. Recommended Tool Compounds: Application of YM-254890 and FR900359 to Interrogate Gα q/11-Mediated Signaling Pathways. ACS Pharmacol Transl Sci 2023; 6:1790-1800. [PMID: 38093837 PMCID: PMC10714435 DOI: 10.1021/acsptsci.3c00214] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2024]
Abstract
The macrocyclic depsipeptides YM-254890 (YM) and FR900359 (FR) are natural products, which inhibit heterotrimeric Gαq/11 proteins with high potency and outstanding selectivity. Historically, pharmacological modulation of Gα proteins was only achieved by treatment with pertussis toxin and cholera toxin, whose application can be tedious and is restricted to the inhibition of Gαi/o proteins and activation of Gαs proteins, respectively. The breakthrough discovery and characterization of YM and FR rendered the closely related Gαq, Gα11, and Gα14 proteins amenable to pharmacological inhibition, and since then, both compounds have become widely used in molecular pharmacology and were also proven to be efficacious in animal models of disease. In the past years, both YM and FR were thoroughly characterized and have substantially contributed to an improved understanding of Gαq/11 signaling on a molecular and cellular level. Yet, the possibilities to interrogate Gαq/11 signaling in complex systems have only been exploited in a very limited number of studies, whose promising initial results warrant further application of YM and FR in basic and translational research. As both compounds have become commercially available as of late, this review focuses on their application in cell-based assays and in vivo systems, highlighting their qualities as tool compounds and providing instructions for their use.
Collapse
Affiliation(s)
- Jan Hendrik Voss
- Department of Physiology and Pharmacology,
Section of Receptor Biology and Signaling, Karolinska Institutet, S-171 65 Stockholm, Sweden
| |
Collapse
|
14
|
Baqai U, Kurimchak AM, Trachtenberg IV, Purwin TJ, Haj JI, Han A, Luo K, Pachon NF, Jeon A, Chua V, Davies MA, Gutkind JS, Benovic JL, Duncan JS, Aplin AE. Kinome profiling identifies MARK3 and STK10 as potential therapeutic targets in uveal melanoma. J Biol Chem 2023; 299:105418. [PMID: 37923138 PMCID: PMC10716579 DOI: 10.1016/j.jbc.2023.105418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 10/05/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Most uveal melanoma cases harbor activating mutations in either GNAQ or GNA11. Despite activation of the mitogen-activated protein kinase (MAPK) signaling pathway downstream of Gαq/11, there are no effective targeted kinase therapies for metastatic uveal melanoma. The human genome encodes numerous understudied kinases, also called the "dark kinome". Identifying additional kinases regulated by Gαq/11 may uncover novel therapeutic targets for uveal melanoma. In this study, we treated GNAQ-mutant uveal melanoma cell lines with a Gαq/11 inhibitor, YM-254890, and conducted a kinase signaling proteomic screen using multiplexed-kinase inhibitors followed by mass spectrometry. We observed downregulated expression and/or activity of 22 kinases. A custom siRNA screen targeting these kinases demonstrated that knockdown of microtubule affinity regulating kinase 3 (MARK3) and serine/threonine kinase 10 (STK10) significantly reduced uveal melanoma cell growth and decreased expression of cell cycle proteins. Additionally, knockdown of MARK3 but not STK10 decreased ERK1/2 phosphorylation. Analysis of RNA-sequencing and proteomic data showed that Gαq signaling regulates STK10 expression and MARK3 activity. Our findings suggest an involvement of STK10 and MARK3 in the Gαq/11 oncogenic pathway and prompt further investigation into the specific roles and targeting potential of these kinases in uveal melanoma.
Collapse
Affiliation(s)
- Usman Baqai
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Alison M Kurimchak
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Isabella V Trachtenberg
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Timothy J Purwin
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jelan I Haj
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Anna Han
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju, Jeollabuk-do, Republic of Korea
| | - Kristine Luo
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Nikole Fandino Pachon
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Angela Jeon
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Vivian Chua
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - J Silvio Gutkind
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - James S Duncan
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Andrew E Aplin
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
15
|
Onken MD, Erdmann-Gilmore P, Zhang Q, Thapa K, King E, Kaltenbronn KM, Noda SE, Makepeace CM, Goldfarb D, Babur Ö, Townsend RR, Blumer KJ. Protein Kinase Signaling Networks Driven by Oncogenic Gq/11 in Uveal Melanoma Identified by Phosphoproteomic and Bioinformatic Analyses. Mol Cell Proteomics 2023; 22:100649. [PMID: 37730182 PMCID: PMC10616553 DOI: 10.1016/j.mcpro.2023.100649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/22/2023] [Accepted: 09/17/2023] [Indexed: 09/22/2023] Open
Abstract
Metastatic uveal melanoma (UM) patients typically survive only 2 to 3 years because effective therapy does not yet exist. Here, to facilitate the discovery of therapeutic targets in UM, we have identified protein kinase signaling mechanisms elicited by the drivers in 90% of UM tumors: mutant constitutively active G protein α-subunits encoded by GNAQ (Gq) or GNA11 (G11). We used the highly specific Gq/11 inhibitor FR900359 (FR) to elucidate signaling networks that drive proliferation, metabolic reprogramming, and dedifferentiation of UM cells. We determined the effects of FR on the proteome and phosphoproteome of UM cells as indicated by bioinformatic analyses with CausalPath and site-specific gene set enrichment analysis. We found that inhibition of oncogenic Gq/11 caused deactivation of PKC, Erk, and the cyclin-dependent kinases CDK1 and CDK2 that drive proliferation. Inhibition of oncogenic Gq/11 in UM cells with low metastatic risk relieved inhibitory phosphorylation of polycomb-repressive complex subunits that regulate melanocytic redifferentiation. Site-specific gene set enrichment analysis, unsupervised analysis, and functional studies indicated that mTORC1 and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 2 drive metabolic reprogramming in UM cells. Together, these results identified protein kinase signaling networks driven by oncogenic Gq/11 that regulate critical aspects of UM cell biology and provide targets for therapeutic investigation.
Collapse
Affiliation(s)
- Michael D Onken
- Department of Biochemistry and Molecular Biophysics, Washington University in St Louis, St Louis, Missouri, USA.
| | | | - Qiang Zhang
- Department of Medicine, Washington University in St Louis, St Louis, Missouri, USA
| | - Kisan Thapa
- Department of Computer Science, University of Massachusetts Boston, Boston, Massachusetts, USA
| | - Emily King
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Kevin M Kaltenbronn
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Sarah E Noda
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Carol M Makepeace
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Dennis Goldfarb
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Özgün Babur
- Department of Computer Science, University of Massachusetts Boston, Boston, Massachusetts, USA
| | - R Reid Townsend
- Department of Medicine, Washington University in St Louis, St Louis, Missouri, USA
| | - Kendall J Blumer
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA.
| |
Collapse
|
16
|
Hanke W, Alenfelder J, Liu J, Gutbrod P, Kehraus S, Crüsemann M, Dörmann P, Kostenis E, Scholz M, König GM. The Bacterial G q Signal Transduction Inhibitor FR900359 Impairs Soil-Associated Nematodes. J Chem Ecol 2023; 49:549-569. [PMID: 37453001 PMCID: PMC10725363 DOI: 10.1007/s10886-023-01442-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023]
Abstract
The cyclic depsipeptide FR900359 (FR) is derived from the soil bacterium Chromobacterium vaccinii and known to bind Gq proteins of mammals and insects, thereby abolishing the signal transduction of their Gq protein-coupled receptors, a process that leads to severe physiological consequences. Due to their highly conserved structure, Gq family of proteins are a superior ecological target for FR producing organisms, resulting in a defense towards a broad range of harmful organisms. Here, we focus on the question whether bacteria like C. vaccinii are important factors in soil in that their secondary metabolites impair, e.g., plant harming organisms like nematodes. We prove that the Gq inhibitor FR is produced under soil-like conditions. Furthermore, FR inhibits heterologously expressed Gαq proteins of the nematodes Caenorhabditis elegans and Heterodera schachtii in the micromolar range. Additionally, in vivo experiments with C. elegans and the plant parasitic cyst nematode H. schachtii demonstrated that FR reduces locomotion of C. elegans and H. schachtii. Finally, egg-laying of C. elegans and hatching of juvenile stage 2 of H. schachtii from its cysts is inhibited by FR, suggesting that FR might reduce nematode dispersion and proliferation. This study supports the idea that C. vaccinii and its excreted metabolome in the soil might contribute to an ecological equilibrium, maintaining and establishing the successful growth of plants.
Collapse
Affiliation(s)
- Wiebke Hanke
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, D-53115, Bonn, Germany
| | - Judith Alenfelder
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, D-53115, Bonn, Germany
| | - Jun Liu
- Neural Information Flow, Max Planck Institute for Neurobiology of Behavior - CAESAR, Ludwig-Erhard-Allee 2, D-53175, Bonn, Germany
| | - Philipp Gutbrod
- Institute of Molecular Physiology and Biotechnology of Plants (IMBIO), University of Bonn, Karlrobert-Kreiten-Straße 13, D-53115, Bonn, Germany
- Bonn International Graduate School - Land and Food, University of Bonn, Katzenburgweg 9, D-53115, Bonn, Germany
| | - Stefan Kehraus
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, D-53115, Bonn, Germany
| | - Max Crüsemann
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, D-53115, Bonn, Germany
| | - Peter Dörmann
- Institute of Molecular Physiology and Biotechnology of Plants (IMBIO), University of Bonn, Karlrobert-Kreiten-Straße 13, D-53115, Bonn, Germany
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, D-53115, Bonn, Germany
| | - Monika Scholz
- Neural Information Flow, Max Planck Institute for Neurobiology of Behavior - CAESAR, Ludwig-Erhard-Allee 2, D-53175, Bonn, Germany
| | - Gabriele M König
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, D-53115, Bonn, Germany.
| |
Collapse
|
17
|
Voss JH, Crüsemann M, Bartling CR, Kehraus S, Inoue A, König GM, Strømgaard K, Müller CE. Structure-affinity and structure-residence time relationships of macrocyclic Gα q protein inhibitors. iScience 2023; 26:106492. [PMID: 37091255 PMCID: PMC10119753 DOI: 10.1016/j.isci.2023.106492] [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: 01/21/2023] [Revised: 02/02/2023] [Accepted: 03/21/2023] [Indexed: 04/25/2023] Open
Abstract
The macrocyclic depsipeptides YM-254890 (YM) and FR900359 (FR) are potent inhibitors of Gαq/11 proteins. They are important pharmacological tools and have potential as therapeutic drugs. The hydrogenated, tritium-labeled YM and FR derivatives display largely different residence times despite similar structures. In the present study we established a competition-association binding assay to determine the dissociation kinetics of unlabeled Gq protein inhibitors. Structure-affinity and structure-residence time relationships were analyzed. Small structural modifications had a large impact on residence time. YM and FR exhibited 4- to 10-fold higher residence times than their hydrogenated derivatives. While FR showed pseudo-irreversible binding, YM displayed much faster dissociation from its target. The isopropyl anchor present in FR and some derivatives was essential for slow dissociation. These data provide a basis for future drug design toward modulating residence times of macrocyclic Gq protein inhibitors, which has been recognized as a crucial determinant for therapeutic outcome.
Collapse
Affiliation(s)
- Jan H. Voss
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Max Crüsemann
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Christian R.O. Bartling
- Department of Drug Design and Pharmacology, Center for Biopharmaceuticals, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Stefan Kehraus
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Asuka Inoue
- Tohoku University, Graduate School of Pharmaceutical Sciences, Sendai, Miyagi 980-8578, Japan
| | - Gabriele M. König
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, Center for Biopharmaceuticals, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Christa E. Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
- Corresponding author
| |
Collapse
|
18
|
Pistorius D, Buntin K, Richard E, Rust M, Bouquet C, Wollbrett S, Weber E, Dietschin D, Bruccoleri R, Oakeley E, Petersen F. Valhidepsin Lipopeptides from Chromobacterium vaccinii: Structures, Biosynthesis, and Coregulation with FR900359 Production. JOURNAL OF NATURAL PRODUCTS 2023; 86:246-255. [PMID: 36745695 DOI: 10.1021/acs.jnatprod.2c00825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Microbial secondary metabolites continue to provide a valuable source of both chemical matter and inspiration for drug discovery in a broad range of therapeutic areas. Beyond this, the corresponding microorganisms represent a sustainable modality for biotechnological production of structurally complex molecules at the quantities required for drug development or even commercial manufacturing. Chromobacterium vaccinii, which has recently been reported as a producer of the pharmacologically highly important Gq inhibitor FR900359 (FR), represents such an example. The characterization of an orphan biosynthetic gene cluster (BGC) located directly downstream of the frs BCG led to the discovery of eight new lipopeptides, valhidepsins A-H (1-8), produced by C. vaccinii. Their chemical structures were elucidated through analysis of 1D and 2D NMR data and high-resolution MS/MS fragmentation methods. The valhidepsins did not display significant antibiotic nor cytotoxic activities but showed surfactant properties. The cluster-compound correlation was demonstrated by generation of a knockout mutant, which abolished production of valhidepsins. This knockout mutant yielded a significantly increased isolated yield of FR.
Collapse
Affiliation(s)
- Dominik Pistorius
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056 Basel, Switzerland
| | - Kathrin Buntin
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056 Basel, Switzerland
| | - Etienne Richard
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056 Basel, Switzerland
| | - Michael Rust
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056 Basel, Switzerland
| | - Caroline Bouquet
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056 Basel, Switzerland
| | - Séverine Wollbrett
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056 Basel, Switzerland
| | - Eric Weber
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056 Basel, Switzerland
| | - Daniele Dietschin
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056 Basel, Switzerland
| | | | - Edward Oakeley
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056 Basel, Switzerland
| | - Frank Petersen
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056 Basel, Switzerland
| |
Collapse
|
19
|
Carvajal RD, Sacco JJ, Jager MJ, Eschelman DJ, Olofsson Bagge R, Harbour JW, Chieng ND, Patel SP, Joshua AM, Piperno-Neumann S. Advances in the clinical management of uveal melanoma. Nat Rev Clin Oncol 2023; 20:99-115. [PMID: 36600005 DOI: 10.1038/s41571-022-00714-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 01/05/2023]
Abstract
Melanomas arising in the uveal tract of the eye are a rare form of the disease with a biology and clinical phenotype distinct from their more common cutaneous counterparts. Treatment of primary uveal melanoma with radiotherapy, enucleation or other modalities achieves local control in more than 90% of patients, although 40% or more ultimately develop distant metastases, most commonly in the liver. Until January 2022, no systemic therapy had received regulatory approval for patients with metastatic uveal melanoma, and these patients have historically had a dismal prognosis owing to the limited efficacy of the available treatments. A series of seminal studies over the past two decades have identified highly prevalent early, tumour-initiating oncogenic genomic aberrations, later recurring prognostic alterations and immunological features that characterize uveal melanoma. These advances have driven the development of a number of novel emerging treatments, including tebentafusp, the first systemic therapy to achieve regulatory approval for this disease. In this Review, our multidisciplinary and international group of authors summarize the biology of uveal melanoma, management of primary disease and surveillance strategies to detect recurrent disease, and then focus on the current standard and emerging regional and systemic treatment approaches for metastatic uveal melanoma.
Collapse
Affiliation(s)
- Richard D Carvajal
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| | - Joseph J Sacco
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - David J Eschelman
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - J William Harbour
- Department of Ophthalmology and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Nicholas D Chieng
- Medical Imaging Services, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Sapna P Patel
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Anthony M Joshua
- Department of Medical Oncology, Kinghorn Cancer Centre, St Vincent's Hospital Sydney and Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,School of Clinical Medicine, UNSW Medicine & Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW, Sydney, New South Wales, Australia
| | | |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Wei AZ, Maniar AB, Carvajal RD. New targeted and epigenetic therapeutic strategies for the treatment of uveal melanoma. Cancer Gene Ther 2022; 29:1819-1826. [PMID: 35236928 DOI: 10.1038/s41417-022-00443-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/14/2022] [Accepted: 02/08/2022] [Indexed: 02/07/2023]
Abstract
Uveal melanoma (UM) is a rare, genetically bland ocular malignancy with excellent local treatment options, but no disease-specific therapies are approved for use in the metastatic setting by the Food and Drug Administration. Metastatic UM (mUM) confers a prognosis of ~15 months. Unlike cutaneous melanoma, UM is poorly responsive to checkpoint inhibitors and cytotoxic chemotherapy highlighting the importance of clarifying vulnerable disease-specific mechanisms, such as cell cycle or metabolic pathways necessary for tumor growth and survival. The elucidation of signaling pathways downstream of the frequently mutated GNA GTPase such as PKC/MAPK/ERK/MEK, PI3K/AKT, and YAP-Hippo have offered potential targets. Potentially druggable epigenetic targets due to BAP1-mutated UM have also been identified, including proteins involved with histone deacetylation and DNA splicing. This review describes the preclinical rationale for the development of targeted therapies and current strategies currently being studied in clinical trials or will be in the near future.
Collapse
Affiliation(s)
- Alexander Z Wei
- Columbia University Irving Medical Center, New York, New York, USA
| | - Ashray B Maniar
- Columbia University Irving Medical Center, New York, New York, USA
| | | |
Collapse
|
23
|
Caksa S, Baqai U, Aplin AE. The future of targeted kinase inhibitors in melanoma. Pharmacol Ther 2022; 239:108200. [PMID: 35513054 PMCID: PMC10187889 DOI: 10.1016/j.pharmthera.2022.108200] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 12/13/2022]
Abstract
Melanoma is a cancer of the pigment-producing cells of the body and its incidence is rising. Targeted inhibitors that act against kinases in the MAPK pathway are approved for BRAF-mutant metastatic cutaneous melanoma and increase patients' survival. Response to these therapies is limited by drug resistance and is less durable than with immune checkpoint inhibition. Conversely, rare melanoma subtypes have few therapeutic options for advanced disease and MAPK pathway targeting agents show minimal anti-tumor effects. Nevertheless, there is a future for targeted kinase inhibitors in melanoma: in new applications such as adjuvant or neoadjuvant therapy and in novel combinations with immunotherapies or other targeted therapies. Pre-clinical studies continue to identify tumor dependencies and their corresponding actionable drug targets, paving the way for rational targeted kinase inhibitor combinations as a personalized medicine approach for melanoma.
Collapse
Affiliation(s)
- Signe Caksa
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Usman Baqai
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
24
|
Park JJ, Stewart A, Irvine M, Pedersen B, Ming Z, Carlino MS, Diefenbach RJ, Rizos H. Protein kinase inhibitor responses in uveal melanoma reflects a diminished dependency on PKC-MAPK signaling. Cancer Gene Ther 2022; 29:1384-1393. [PMID: 35352024 PMCID: PMC9576594 DOI: 10.1038/s41417-022-00457-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/20/2022] [Accepted: 03/04/2022] [Indexed: 01/20/2023]
Abstract
Uveal melanoma (UM) is a rare cancer arising from melanocytes in the uveal tract of the eye. Despite effective primary treatment, there is no approved therapy for metastatic UM and prognosis and survival remain poor. Over 90% of UM are driven by mutations affecting the Gα subunits encoded by the GNAQ and GNA11 genes. These mutations activate downstream and targetable signaling pathways, including the protein kinase C (PKC) cascade. PKC inhibitors have been used in clinical trials for metastatic UM but have shown limited efficacy. In this study, we examined the signaling and functional effects of two PKC inhibitors (AEB071 and IDE196) in a panel of UM cell models. In response to PKC inhibition, all UM cell lines showed potent suppression of PKC activity, but this was not sufficient to predict PKC inhibitor sensitivity and only two UM cell lines showed substantial PKC inhibitor-induced cell death. The differences in UM cell responses to PKC inhibition were not attributable to the degree or timing of PKC suppression or inhibition of the downstream mitogen-activated protein kinase (MAPK) or phosphatidylinositol-3-kinase (PI3K) pathways. Instead, UM cell show complex, PKC-independent signaling pathways that contribute to their survival and resistance to targeted therapies.
Collapse
Affiliation(s)
- John J Park
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Melanoma Institute Australia, Sydney, Australia
| | - Ashleigh Stewart
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Mal Irvine
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Bernadette Pedersen
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Zizhen Ming
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, Sydney, Australia
- Department of Medical Oncology, Westmead and Blacktown Hospitals, Sydney, Australia
| | - Russell J Diefenbach
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Melanoma Institute Australia, Sydney, Australia
| | - Helen Rizos
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia.
- Melanoma Institute Australia, Sydney, Australia.
| |
Collapse
|
25
|
Randolph CE, Dwyer MB, Aumiller JL, Dixon AJ, Inoue A, Osei-Owusu P, Wedegaertner PB. Enhanced membrane binding of oncogenic G protein αqQ209L confers resistance to inhibitor YM-254890. J Biol Chem 2022; 298:102538. [PMID: 36174676 PMCID: PMC9626947 DOI: 10.1016/j.jbc.2022.102538] [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: 03/24/2022] [Revised: 09/15/2022] [Accepted: 09/18/2022] [Indexed: 11/29/2022] Open
Abstract
Heterotrimeric G proteins couple activated G protein–coupled receptors (GPCRs) to intracellular signaling pathways. They can also function independently of GPCR activation upon acquiring mutations that prevent GTPase activity and result in constitutive signaling, as occurs with the αqQ209L mutation in uveal melanoma. YM-254890 (YM) can inhibit signaling by both GPCR-activated WT αq and GPCR-independent αqQ209L. Although YM inhibits WT αq by binding to αq-GDP and preventing GDP/GTP exchange, the mechanism of YM inhibition of cellular αqQ209L remains to be fully understood. Here, we show that YM promotes a subcellular redistribution of αqQ209L from the plasma membrane (PM) to the cytoplasm. To test if this loss of PM localization could contribute to the mechanism of inhibition of αqQ209L by YM, we developed and examined N-terminal mutants of αqQ209L, termed PM-restricted αqQ209L, in which the addition of membrane-binding motifs enhanced PM localization and prevented YM-promoted redistribution. Treatment of cells with YM failed to inhibit signaling by these PM-restricted αqQ209L. Additionally, pull-down experiments demonstrated that YM promotes similar conformational changes in both αqQ209L and PM-restricted αqQ209L, resulting in increased binding to βγ and decreased binding to regulator RGS2, and effectors p63RhoGEF-DH/PH and phospholipase C-β. GPCR-dependent signaling by PM-restricted WT αq is strongly inhibited by YM, demonstrating that resistance to YM inhibition by membrane-binding mutants is specific to constitutively active αqQ209L. Together, these results indicate that changes in membrane binding impact the ability of YM to inhibit αqQ209L and suggest that YM contributes to inhibition of αqQ209L by promoting its relocalization.
Collapse
Affiliation(s)
- Clinita E Randolph
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Morgan B Dwyer
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Jenna L Aumiller
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Alethia J Dixon
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Patrick Osei-Owusu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Philip B Wedegaertner
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA.
| |
Collapse
|
26
|
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: 1.7] [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
|
27
|
Mukherjee N, Dart CR, Amato CM, Honig-Frand A, Lambert JR, Lambert KA, Robinson WA, Tobin RP, McCarter MD, Couts KL, Fujita M, Norris DA, Shellman YG. Expression Differences in BCL2 Family Members between Uveal and Cutaneous Melanomas Account for Varying Sensitivity to BH3 Mimetics. J Invest Dermatol 2022; 142:1912-1922.e7. [PMID: 34942200 PMCID: PMC9635014 DOI: 10.1016/j.jid.2021.11.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 11/04/2021] [Accepted: 11/19/2021] [Indexed: 10/19/2022]
Abstract
Uveal melanoma (UM) is a subtype of melanoma. Although they share a melanocytic origin with cutaneous melanoma (CM), patients with UM have few treatment options. BCL2 homologous 3 mimetics are small-molecule drugs that mimic proapoptotic BCL2 family members. We compared BCL2 family member expression between UM and CM using immunoblot and The Cancer Genome Atlas transcriptomic analysis. UM has a unique signature of low BFL1 and high PUMA proteins compared with CM and 30 other cancer types, making them an attractive candidate for BCL2 homologous 3 protein mimetics. We tested the efficacy of a BCL2 inhibitor and MCL1 inhibitor (MCL1i) in UM, with viability assays, live-cell imaging, sphere assays, and mouse xenograft models. UM had a higher sensitivity to MCL1i than CM. Overexpression of BFL1 or knockdown of PUMA made the UM more resistant to MCL1i. In contrast, MAPK/extracellular signal‒regulated kinase inhibitor treatment in CM made them more sensitive to MCL1i. However, MCL1i-alone treatment was not very effective to reduce the UM initiating cells; to overcome this, we employed a combination of MCL1i with BCL2 inhibitor that synergistically inhibited UM initiating cell's capacity to expand. Overall, we identify a distinct expression profile of BCL2 family members for UM that makes them susceptible to BCL2 homologous 3 mimetics.
Collapse
Affiliation(s)
- Nabanita Mukherjee
- Department of Dermatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Chiara R Dart
- Department of Dermatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Carol M Amato
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Adam Honig-Frand
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - James R Lambert
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Karoline A Lambert
- Department of Dermatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - William A Robinson
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard P Tobin
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | - Martin D McCarter
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kasey L Couts
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mayumi Fujita
- Department of Dermatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Dermatology Section, U.S. Department of Veterans Affairs Medical Center, Denver, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David A Norris
- Department of Dermatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Dermatology Section, U.S. Department of Veterans Affairs Medical Center, Denver, Colorado, USA
| | - Yiqun G Shellman
- Department of Dermatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
28
|
Silva-Rodríguez P, Fernández-Díaz D, Bande M, Pardo M, Loidi L, Blanco-Teijeiro MJ. GNAQ and GNA11 Genes: A Comprehensive Review on Oncogenesis, Prognosis and Therapeutic Opportunities in Uveal Melanoma. Cancers (Basel) 2022; 14:3066. [PMID: 35804836 PMCID: PMC9264989 DOI: 10.3390/cancers14133066] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 11/17/2022] Open
Abstract
The GNAQ and GNA11 genes are mutated in almost 80-90% of uveal melanomas in a mutually exclusive pattern. These genes encode the alpha subunits of the heterotrimeric G proteins, Gq and G11; thus, mutations of these genes result in the activation of several important signaling pathways, including phospholipase C, and activation of the transcription factor YAP. It is well known that both of them act as driver genes in the oncogenic process and it has been assumed that they do not play a role in the prognosis of these tumours. However, it has been hypothesised that mutations in these genes could give rise to molecularly and clinically distinct types of uveal melanomas. It has also been questioned whether the type and location of mutation in the GNAQ and GNA11 genes may affect the progression of these tumours. All of these questions, except for their implications in carcinogenesis, remain controversial. Uveal melanoma has a distinctive genetic profile, and specific recurrent mutations, which make it a potential candidate for treatment with targeted therapy. Given that the most frequent mutations are those observed in the GNAQ and GNA11 genes, and that both genes are involved in oncogenesis, these molecules, as well as the downstream signalling pathways in which they are involved, have been proposed as promising potential therapeutic targets. Therefore, in this review, special attention is paid to the current data related to the possible prognostic implications of both genes from different perspectives, as well as the therapeutic options targeting them.
Collapse
Affiliation(s)
- Paula Silva-Rodríguez
- Fundación Pública Galega de Medicina Xenómica, Clinical University Hospital, SERGAS, 15706 Santiago de Compostela, Spain;
- Tumores Intraoculares en el Adulto, Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain; (D.F.-D.); (M.B.); (M.J.B.-T.)
| | - Daniel Fernández-Díaz
- Tumores Intraoculares en el Adulto, Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain; (D.F.-D.); (M.B.); (M.J.B.-T.)
| | - Manuel Bande
- Tumores Intraoculares en el Adulto, Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain; (D.F.-D.); (M.B.); (M.J.B.-T.)
- Department of Ophthalmology, University Hospital of Santiago de Compostela, Ramon Baltar S/N, 15706 Santiago de Compostela, Spain
| | - María Pardo
- Grupo Obesidómica, Instituto de Investigación Sanitaria de Santiago (IDIS), CIBEROBN, ISCIII, 15706 Santiago de Compostela, Spain;
| | - Lourdes Loidi
- Fundación Pública Galega de Medicina Xenómica, Clinical University Hospital, SERGAS, 15706 Santiago de Compostela, Spain;
| | - María José Blanco-Teijeiro
- Tumores Intraoculares en el Adulto, Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain; (D.F.-D.); (M.B.); (M.J.B.-T.)
- Department of Ophthalmology, University Hospital of Santiago de Compostela, Ramon Baltar S/N, 15706 Santiago de Compostela, Spain
| |
Collapse
|
29
|
Baqai U, Purwin TJ, Bechtel N, Chua V, Han A, Hartsough EJ, Kuznetsoff JN, Harbour JW, Aplin AE. Multi-omics profiling shows BAP1 loss is associated with upregulated cell adhesion molecules in uveal melanoma. Mol Cancer Res 2022; 20:1260-1271. [DOI: 10.1158/1541-7786.mcr-21-0657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 02/04/2022] [Accepted: 04/11/2022] [Indexed: 11/16/2022]
Abstract
Abstract
BRCA1-associated protein 1 (BAP1) is a tumor suppressor gene that is mutated in cancer, including uveal melanoma (UM). Loss-of-function BAP1 mutations are associated with UM metastasis and poor prognosis, but the mechanisms underlying these effects remain unclear. Upregulation of cell-cell adhesion proteins is involved with collective migration and metastatic seeding of cancer cells. Here, we show that BAP1 loss in UM patient samples is associated with upregulated gene expression of multiple cell adhesion molecules (CAMs), including E-cadherin (CDH1), cell adhesion molecule 1 (CADM1), and syndecan-2 (SDC2). Similar findings were observed in UM cell lines and scRNA seq data from UM patient samples. BAP1 re-expression in UM cells reduced E-cadherin and CADM1 levels. Functionally, knockdown of E-cadherin decreased spheroid cluster formation and knockdown of CADM1 decreased growth of BAP1 mutant UM cells. Together, our findings demonstrate that BAP1 regulates the expression of CAMs which may regulate metastatic traits. Implications: BAP1 mutations and increased metastasis may be due to upregulation of cell adhesion molecules.
Collapse
Affiliation(s)
- Usman Baqai
- Thomas Jefferson University, Philadelphia, PA, United States
| | | | - Nelisa Bechtel
- Thomas Jefferson University, Philadelphia, PA, United States
| | - Vivian Chua
- Thomas Jefferson University, Philadelphia, PA, United States
| | - Anna Han
- Thomas Jefferson University, Philadelphia, PA, United States
| | - Edward J. Hartsough
- Drexel University College of Medicine, Philadelphia, Pennsylvania, United States
| | | | | | - Andrew E. Aplin
- Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
30
|
Han A, Chua V, Baqai U, Purwin TJ, Bechtel N, Hunter E, Tiago M, Seifert E, Speicher DW, Schug ZT, Harbour JW, Aplin AE. Pyruvate dehydrogenase inactivation causes glycolytic phenotype in BAP1 mutant uveal melanoma. Oncogene 2022; 41:1129-1139. [PMID: 35046531 PMCID: PMC9066178 DOI: 10.1038/s41388-021-02154-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 12/02/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022]
Abstract
Effective therapeutic options are still lacking for uveal melanoma (UM) patients who develop metastasis. Metastatic traits of UM are linked to BRCA1-associated protein 1 (BAP1) mutations. Cell metabolism is re-programmed in UM with BAP1 mutant UM, but the underlying mechanisms and opportunities for therapeutic intervention remain unclear. BAP1 mutant UM tumors have an elevated glycolytic gene expression signature, with increased expression of pyruvate dehydrogenase (PDH) complex and PDH kinase (PDHK1). Furthermore, BAP1 mutant UM cells showed higher levels of phosphorylated PDHK1 and PDH that was associated with an upregulated glycolytic profile compared to BAP1 wild-type UM cells. Suppressing PDHK1-PDH phosphorylation decreased glycolytic capacity and cell growth, and induced cell cycle arrest of BAP1 mutant UM cells. Our results suggest that PDHK1-PDH phosphorylation is a causative factor of glycolytic phenotypes found in BAP1 mutant UM and propose a therapeutic opportunity for BAP1 mutant UM patients.
Collapse
Affiliation(s)
- Anna Han
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
| | - Vivian Chua
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Usman Baqai
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Timothy J Purwin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Nelisa Bechtel
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Emily Hunter
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Manoela Tiago
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Erin Seifert
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - David W Speicher
- Proteomics and Metabolomics Facility, The Wistar Institute, Philadelphia, PA, 19104, USA
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Zachary T Schug
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - J William Harbour
- Bascom Palmer Eye Institute, Sylvester Comprehensive Cancer Center and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, 33146, USA
- Department of Ophthalmology, Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| |
Collapse
|
31
|
OUP accepted manuscript. J Pharm Pharmacol 2022; 74:660-680. [DOI: 10.1093/jpp/rgac009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/02/2022] [Indexed: 11/12/2022]
|
32
|
Bolger GB. The cAMP-signaling cancers: Clinically-divergent disorders with a common central pathway. Front Endocrinol (Lausanne) 2022; 13:1024423. [PMID: 36313756 PMCID: PMC9612118 DOI: 10.3389/fendo.2022.1024423] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/27/2022] [Indexed: 12/01/2022] Open
Abstract
The cAMP-signaling cancers, which are defined by functionally-significant somatic mutations in one or more elements of the cAMP signaling pathway, have an unexpectedly wide range of cell origins, clinical manifestations, and potential therapeutic options. Mutations in at least 9 cAMP signaling pathway genes (TSHR, GPR101, GNAS, PDE8B, PDE11A, PRKARA1, PRKACA, PRKACB, and CREB) have been identified as driver mutations in human cancer. Although all cAMP-signaling pathway cancers are driven by mutation(s) that impinge on a single signaling pathway, the ultimate tumor phenotype reflects interactions between five critical variables: (1) the precise gene(s) that undergo mutation in each specific tumor type; (2) the effects of specific allele(s) in any given gene; (3) mutations in modifier genes (mutational "context"); (4) the tissue-specific expression of various cAMP signaling pathway elements in the tumor stem cell; and (5) and the precise biochemical regulation of the pathway components in tumor cells. These varying oncogenic mechanisms reveal novel and important targets for drug discovery. There is considerable diversity in the "druggability" of cAMP-signaling components, with some elements (GPCRs, cAMP-specific phosphodiesterases and kinases) appearing to be prime drug candidates, while other elements (transcription factors, protein-protein interactions) are currently refractory to robust drug-development efforts. Further refinement of the precise driver mutations in individual tumors will be essential for directing priorities in drug discovery efforts that target these mutations.
Collapse
|
33
|
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: 2.5] [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
|
34
|
Pistorius D, Buntin K, Weber E, Richard E, Bouquet C, Wollbrett S, Regenass H, Peón V, Böhm M, Kessler R, Gempeler T, Haberkorn A, Wimmer L, Lanshoeft C, Davis J, Hainzl D, D'Alessio JA, Manchado E, Petersen F. Promoter-Driven Overexpression in Chromobacterium vaccinii Facilitates Access to FR900359 and Yields Novel Low Abundance Analogs. Chemistry 2021; 28:e202103888. [PMID: 34878202 DOI: 10.1002/chem.202103888] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Indexed: 11/11/2022]
Abstract
Access to the cyclic depsipeptide FR900359 (FR), a selective Gq/11 protein inhibitor of high pharmacological interest and a potential lead molecule for targeted therapy of cancers with oncogenic GNAQ or GNA11 mutations (encoding Gq and G11 respectively), has been challenging ever since its initial discovery more than three decades ago. The recent discovery of Chromobacterium vaccinii as a cultivable FR producer enables the development of approaches leading to a high-yielding, scalable and sustainable biotechnological process for production of FR, thereby removing this bottleneck. Here we characterize different promoters in exchange of the native promoter of the FR assembly line, resulting in an overexpression mutant with significantly increased production of FR. Thereby, the isolation and structure elucidation of novel FR analogs of low abundance is enabled. Further, we explore the antiproliferative activities of fifteen chromodepsins against uveal melanoma cell lines harboring Gq/11 mutations and characterize the major metabolite of FR formed in plasma.
Collapse
Affiliation(s)
- Dominik Pistorius
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Kathrin Buntin
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Eric Weber
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Etienne Richard
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Caroline Bouquet
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Séverine Wollbrett
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Hugo Regenass
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Victor Peón
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Marcel Böhm
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Régis Kessler
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Thomas Gempeler
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Anne Haberkorn
- Oncology Disease Area, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Laurin Wimmer
- Chemical & Analytical Development, Technical Research & Development, Global Drug Development, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Christian Lanshoeft
- Pharmakokinetic Sciences, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - John Davis
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, Novartis Pharma AG, 250 Massachusetts Ave, Cambridge, MA, 02139, USA
| | - Dominik Hainzl
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, Novartis Pharma AG, 250 Massachusetts Ave, Cambridge, MA, 02139, USA
| | - Joseph Anthony D'Alessio
- Oncology Disease Area, Novartis Institutes for BioMedical Research, Novartis Pharma AG, 250 Massachusetts Ave, Cambridge, MA, 02139, USA
| | - Eusebio Manchado
- Oncology Disease Area, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Frank Petersen
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| |
Collapse
|
35
|
Rossi E, Croce M, Reggiani F, Schinzari G, Ambrosio M, Gangemi R, Tortora G, Pfeffer U, Amaro A. Uveal Melanoma Metastasis. Cancers (Basel) 2021; 13:5684. [PMID: 34830841 PMCID: PMC8616038 DOI: 10.3390/cancers13225684] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023] Open
Abstract
Uveal melanoma (UM) is characterized by relatively few, highly incident molecular alterations and their association with metastatic risk is deeply understood. Nevertheless, this knowledge has so far not led to innovative therapies for the successful treatment of UM metastases or for adjuvant therapy, leaving survival after diagnosis of metastatic UM almost unaltered in decades. The driver mutations of UM, mainly in the G-protein genes GNAQ and GNA11, activate the MAP-kinase pathway as well as the YAP/TAZ pathway. At present, there are no drugs that target the latter and this likely explains the failure of mitogen activated kinase kinase inhibitors. Immune checkpoint blockers, despite the game changing effect in cutaneous melanoma (CM), show only limited effects in UM probably because of the low mutational burden of 0.5 per megabase and the unavailability of antibodies targeting the main immune checkpoint active in UM. The highly pro-tumorigenic microenvironment of UM also contributes to therapy resistance. However, T-cell redirection by a soluble T-cell receptor that is fused to an anti-CD3 single-chain variable fragment, local, liver specific therapy, new immune checkpoint blockers, and YAP/TAZ specific drugs give new hope to repeating the success of innovative therapy obtained for CM.
Collapse
Affiliation(s)
- Ernesto Rossi
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (E.R.); (G.S.); (G.T.)
| | - Michela Croce
- Laboratory of Biotherapies, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.C.); (R.G.)
| | - Francesco Reggiani
- Laboratory of Epigenetics, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.R.); (M.A.); (A.A.)
| | - Giovanni Schinzari
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (E.R.); (G.S.); (G.T.)
- Medical Oncology, Università Cattolica del S. Cuore, 00168 Rome, Italy
| | - Marianna Ambrosio
- Laboratory of Epigenetics, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.R.); (M.A.); (A.A.)
| | - Rosaria Gangemi
- Laboratory of Biotherapies, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.C.); (R.G.)
| | - Giampaolo Tortora
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (E.R.); (G.S.); (G.T.)
- Medical Oncology, Università Cattolica del S. Cuore, 00168 Rome, Italy
| | - Ulrich Pfeffer
- Laboratory of Epigenetics, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.R.); (M.A.); (A.A.)
| | - Adriana Amaro
- Laboratory of Epigenetics, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.R.); (M.A.); (A.A.)
| |
Collapse
|
36
|
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: 20] [Impact Index Per Article: 5.0] [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
|
37
|
Chua V, Han A, Bechtel N, Purwin TJ, Hunter E, Liao C, Harbour JW, Aplin AE. The AMP-dependent kinase pathway is upregulated in BAP1 mutant uveal melanoma. Pigment Cell Melanoma Res 2021; 35:78-87. [PMID: 34347929 DOI: 10.1111/pcmr.13007] [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: 09/08/2020] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 02/03/2023]
Abstract
Metastatic uveal melanoma (UM) responds poorly to targeted therapies and immune checkpoint inhibitors. Loss of BRCA1-associated protein 1 (BAP1) via inactivating mutations in the BAP1 gene is associated with UM progression. Thus, molecular alterations caused by BAP1 dysfunction may be novel therapeutic targets for metastatic UM. Here, we found that phosphorylation of AMP-dependent kinase (AMPK) was elevated in BAP1-altered (or mutant) compared to BAP1-unaltered (or wild-type [WT]) UM tumors. As a readout of AMPK pathway activation, phosphorylation of an AMPK downstream effector, acetyl-CoA-carboxylase (ACC), was also elevated. BAP1 re-expression in BAP1-null UM cell lines decreased phospho-AMPK (pAMPK) and phospho-ACC (pACC) levels. AMPK phosphorylation is mediated by calcium/calmodulin dependent protein kinase kinase 2 (CaMKK2) and potentially liver kinase B1 (LKB1) in BAP1 mutant UM cells. Knockdown of AMPKα1/2 reduced the viability of BAP1 mutant UM cells, indicating a survival function of AMPK in BAP1 mutant UM. Our data suggest that the AMPK pathway is an important mechanism mediating the survival of BAP1 mutant UM. Targeting the AMPK pathway may be a novel therapeutic strategy for metastatic UM.
Collapse
Affiliation(s)
- Vivian Chua
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Anna Han
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Nelisa Bechtel
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Timothy J Purwin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Emily Hunter
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Connie Liao
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - J William Harbour
- Bascom Palmer Eye Institute, Sylvester Comprehensive Cancer Center and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA.,Sidney Kimmel Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
38
|
Hermes C, König GM, Crüsemann M. The chromodepsins - chemistry, biology and biosynthesis of a selective Gq inhibitor natural product family. Nat Prod Rep 2021; 38:2276-2292. [PMID: 33998635 DOI: 10.1039/d1np00005e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Covering: up to April 2021The bacterial cyclic depsipeptides FR900359 (FR) and YM-254890 (YM) were shown to selectively inhibit Gαq proteins with high potency and selectivity and have recently emerged as valuable pharmacological tools due to their effective mechanism of action. Here, we summarize important aspects of this small and specialized natural product family, for which we propose the name chromodepsins, starting from their discovery, producing organisms and structural variety. We then review biosynthesis, structure-activity relationships and ecological and evolutionary aspects of the chromodepsins. Lastly, we discuss their mechanism of action, potential medicinal applications and future opportunities and challenges for further use and development of these complex inhibitor molecules from nature.
Collapse
Affiliation(s)
- Cornelia Hermes
- Institute of Pharmaceutical Biology, Rheinische Friedrich-Wilhelms-University of Bonn, 53115 Bonn, Germany.
| | - Gabriele M König
- Institute of Pharmaceutical Biology, Rheinische Friedrich-Wilhelms-University of Bonn, 53115 Bonn, Germany.
| | - Max Crüsemann
- Institute of Pharmaceutical Biology, Rheinische Friedrich-Wilhelms-University of Bonn, 53115 Bonn, Germany.
| |
Collapse
|
39
|
Hitchman TD, Bayshtok G, Ceraudo E, Moore AR, Lee C, Jia R, Wang N, Pachai MR, Shoushtari AN, Francis JH, Guan Y, Chen J, Chang MT, Taylor BS, Sakmar TP, Huber T, Chi P, Chen Y. Combined Inhibition of Gα q and MEK Enhances Therapeutic Efficacy in Uveal Melanoma. Clin Cancer Res 2021; 27:1476-1490. [PMID: 33229459 PMCID: PMC8086191 DOI: 10.1158/1078-0432.ccr-20-2860] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/03/2020] [Accepted: 11/18/2020] [Indexed: 02/03/2023]
Abstract
PURPOSE All uveal melanoma and a fraction of other melanoma subtypes are driven by activation of the G-protein alpha-q (Gαq) pathway. Targeting these melanomas has proven difficult despite advances in the molecular understanding of key driver signaling pathways in the disease pathogenesis. Inhibitors of Gαq have shown promising preclinical results, but their therapeutic activity in distinct Gαq mutational contexts and in vivo have remained elusive. EXPERIMENTAL DESIGN We used an isogenic melanocytic cellular system to systematically examine hotspot mutations in GNAQ (e.g., G48V, R183Q, Q209L) and CYSLTR2 (L129Q) found in human uveal melanoma. This cellular system and human uveal melanoma cell lines were used in vitro and in in vivo xenograft studies to assess the efficacy of Gαq inhibition as a single agent and in combination with MEK inhibition. RESULTS We demonstrate that the Gαq inhibitor YM-254890 inhibited downstream signaling and in vitro growth in all mutants. In vivo, YM-254890 slowed tumor growth but did not cause regression in human uveal melanoma xenografts. Through comprehensive transcriptome analysis, we observed that YM-254890 caused inhibition of the MAPK signaling with evidence of rebound by 24 hours and combination treatment of YM-254890 and a MEK inhibitor led to sustained MAPK inhibition. We further demonstrated that the combination caused synergistic growth inhibition in vitro and tumor shrinkage in vivo. CONCLUSIONS These data suggest that the combination of Gαq and MEK inhibition provides a promising therapeutic strategy and improved therapeutic window of broadly targeting Gαq in uveal melanoma.See related commentary by Neelature Sriramareddy and Smalley, p. 1217.
Collapse
Affiliation(s)
- Tyler D Hitchman
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gabriella Bayshtok
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Emilie Ceraudo
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York
| | - Amanda R Moore
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, New York
| | - Cindy Lee
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ruobing Jia
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, P.R. China
| | - Naitao Wang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mohini R Pachai
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander N Shoushtari
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, New York
| | - Jasmine H Francis
- Opthalmic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Youxin Guan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Juliet Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Matthew T Chang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California
| | - Barry S Taylor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Solna, Sweden
| | - Thomas Huber
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York
| | - Ping Chi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, New York
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, New York
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, New York
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, New York
| |
Collapse
|
40
|
Patt J, Alenfelder J, Pfeil EM, Voss JH, Merten N, Eryilmaz F, Heycke N, Rick U, Inoue A, Kehraus S, Deupi X, Müller CE, König GM, Crüsemann M, Kostenis E. An experimental strategy to probe Gq contribution to signal transduction in living cells. J Biol Chem 2021; 296:100472. [PMID: 33639168 PMCID: PMC8024710 DOI: 10.1016/j.jbc.2021.100472] [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: 12/04/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Heterotrimeric G protein subunits Gαq and Gα11 are inhibited by two cyclic depsipeptides, FR900359 (FR) and YM-254890 (YM), both of which are being used widely to implicate Gq/11 proteins in the regulation of diverse biological processes. An emerging major research question therefore is whether the cellular effects of both inhibitors are on-target, that is, mediated via specific inhibition of Gq/11 proteins, or off-target, that is, the result of nonspecific interactions with other proteins. Here we introduce a versatile experimental strategy to discriminate between these possibilities. We developed a Gαq variant with preserved catalytic activity, but refractory to FR/YM inhibition. A minimum of two amino acid changes were required and sufficient to achieve complete inhibitor resistance. We characterized the novel mutant in HEK293 cells depleted by CRISPR–Cas9 of endogenous Gαq and Gα11 to ensure precise control over the Gα-dependent cellular signaling route. Using a battery of cellular outcomes with known and concealed Gq contribution, we found that FR/YM specifically inhibited cellular signals after Gαq introduction via transient transfection. Conversely, both inhibitors were inert across all assays in cells expressing the drug-resistant variant. These findings eliminate the possibility that inhibition of non-Gq proteins contributes to the cellular effects of the two depsipeptides. We conclude that combined application of FR or YM along with the drug-resistant Gαq variant is a powerful in vitro strategy to discern on-target Gq against off-target non-Gq action. Consequently, it should be of high value for uncovering Gq input to complex biological processes with high accuracy and the requisite specificity.
Collapse
Affiliation(s)
- Julian Patt
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Judith Alenfelder
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Eva Marie Pfeil
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Jan Hendrik Voss
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Nicole Merten
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Funda Eryilmaz
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Nina Heycke
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Uli Rick
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Stefan Kehraus
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Xavier Deupi
- Laboratory of Biomolecular Research and Condensed Matter Theory Group, Paul Scherrer Institute, Villigen, Switzerland
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Gabriele M König
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Max Crüsemann
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany.
| |
Collapse
|
41
|
Schlegel JG, Tahoun M, Seidinger A, Voss JH, Kuschak M, Kehraus S, Schneider M, Matthey M, Fleischmann BK, König GM, Wenzel D, Müller CE. Macrocyclic Gq Protein Inhibitors FR900359 and/or YM-254890-Fit for Translation? ACS Pharmacol Transl Sci 2021; 4:888-897. [PMID: 33860209 DOI: 10.1021/acsptsci.1c00021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Indexed: 12/11/2022]
Abstract
Guanine nucleotide-binding proteins (G proteins) transduce extracellular signals received by G protein-coupled receptors (GPCRs) to intracellular signaling cascades. While GPCRs represent the largest class of drug targets, G protein inhibition has only recently been recognized as a novel strategy for treating complex diseases such as asthma, inflammation, and cancer. The structurally similar macrocyclic depsipeptides FR900359 (FR) and YM-254890 (YM) are potent selective inhibitors of the Gq subfamily of G proteins. FR and YM differ in two positions, FR being more lipophilic than YM. Both compounds are utilized as pharmacological tools to block Gq proteins in vitro and in vivo. However, no detailed characterization of FR and YM has been performed, which is a prerequisite for the compounds' translation into clinical application. Here, we performed a thorough study of both compounds' physicochemical, pharmacokinetic, and pharmacological properties. Chemical stability was high across a large range of pH values, with FR being somewhat more stable than YM. Oral bioavailability and brain penetration of both depsipeptides were low. FR showed lower plasma protein binding and was metabolized significantly faster than YM by human and mouse liver microsomes. FR accumulated in lung after chronic intratracheal or intraperitoneal application, while YM was more distributed to other organs. Most strikingly, the previously observed longer residence time of FR resulted in a significantly prolonged pharmacologic effect as compared to YM in a methacholine-induced bronchoconstriction mouse model. These results prove that changes within a molecule which seem marginal compared to its structural complexity can lead to crucial pharmacological differences.
Collapse
Affiliation(s)
- Jonathan G Schlegel
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Mariam Tahoun
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Alexander Seidinger
- Department of Systems Physiology, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Jan H Voss
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Markus Kuschak
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Stefan Kehraus
- Institute for Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany
| | - Marion Schneider
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Michaela Matthey
- Department of Systems Physiology, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Bernd K Fleischmann
- Institute of Physiology I, Life & Brain Center, Medical Faculty, University of Bonn, 53105 Bonn, Germany
| | - Gabriele M König
- Institute for Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany
| | - Daniela Wenzel
- Department of Systems Physiology, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany.,Institute of Physiology I, Life & Brain Center, Medical Faculty, University of Bonn, 53105 Bonn, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| |
Collapse
|
42
|
Targeting primary and metastatic uveal melanoma with a G protein inhibitor. J Biol Chem 2021; 296:100403. [PMID: 33577798 PMCID: PMC7948511 DOI: 10.1016/j.jbc.2021.100403] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/25/2021] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
Uveal melanoma (UM) is the most common intraocular tumor in adults. Nearly half of UM patients develop metastatic disease and often succumb within months because effective therapy is lacking. A novel therapeutic approach has been suggested by the discovery that UM cell lines driven by mutant constitutively active Gq or G11 can be targeted by FR900359 (FR) or YM-254890, which are bioavailable, selective inhibitors of the Gq/11/14 subfamily of heterotrimeric G proteins. Here, we have addressed the therapeutic potential of FR for UM. We found that FR inhibited all oncogenic Gq/11 mutants reported in UM. FR arrested growth of all Gq/11-driven UM cell lines tested, but induced apoptosis only in a few. Similarly, FR inhibited growth of, but did not efficiently kill, UM tumor cells from biopsies of primary or metastatic tumors. FR evoked melanocytic redifferentiation of UM tumor cells with low (class 1), but not high (class 2), metastatic potential. FR administered systemically below its LD50 strongly inhibited growth of PDX-derived class 1 and class 2 UM tumors in mouse xenograft models and reduced blood pressure transiently. FR did not regress xenografted UM tumors or significantly affect heart rate, liver function, hematopoiesis, or behavior. These results indicated the existence of a therapeutic window in which FR can be explored for treating UM and potentially other diseases caused by constitutively active Gq/11.
Collapse
|
43
|
Guo Z, He X, Jiang C, Shi Y, Zhou N. Activation of Bombyx mori neuropeptide G protein-coupled receptor A19 by neuropeptide RYamides couples to G q protein-dependent signaling pathways. J Cell Biochem 2021; 122:456-471. [PMID: 33399233 DOI: 10.1002/jcb.29874] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022]
Abstract
RYamides constitute a novel family of neuropeptides newly identified in insects, and play important roles in regulating a variety of physiological processes. However, the signaling characteristics and physiological actions of RYamide signaling system remain largely unknown. In the present study, we cloned the full-length complementary DNA of the RYamide receptor BNGR-A19 from Bombyx mori larvae. After expression in mammalian HEK293T and insect Sf9 cells, functional assays revealed that BNGR-A19 was activated by synthetic RYamide peptides, triggering a significant increase in cAMP-response element controlled luciferase activity and Ca2+ mobilization in a Gq inhibitor-sensitive manner. Upon activation by RYamide peptides, BNGR-A19 elicited ERK1/2 phosphorylation via a Gq -PLC-PKC pathway, and also underwent a rapid internalization from the cell surface to the cytoplasm. Further cross-activity analysis indicated that BNGR-A19 exhibited very weak response upon stimulation by high concentration (1 μM) of Bombyx sulfakinin-1, neuropeptide F-1, and short neuropeptide F-1, and vice versa, Bombyx RYamides also showed slight potency for activating Bombyx NPF receptor (BNGR-A4) and sNPF receptor (BNGR-A11). In addition, the quantitative reverse-transcription polymerase chain reaction results showed that the high-level expression of BNGR-A19 was detected in the hindgut and testis, suggesting that the RYamide signaling is likely involved in the regulation of feeding, water homeostasis and testis development. This study provides the first in-depth information on the insect RYamide signaling system, facilitating the further clarification of its endocrinological roles in insect physiology.
Collapse
Affiliation(s)
- Zhiqiang Guo
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Zhejiang, Hangzhou, China
| | - Xiaobai He
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, China
| | - Chaohui Jiang
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Zhejiang, Hangzhou, China
| | - Ying Shi
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Zhejiang, Hangzhou, China
| | - Naiming Zhou
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Zhejiang, Hangzhou, China
| |
Collapse
|
44
|
Functional characterization of uveal melanoma oncogenes. Oncogene 2020; 40:806-820. [PMID: 33262460 PMCID: PMC7856047 DOI: 10.1038/s41388-020-01569-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/05/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023]
Abstract
Uveal melanoma (UM) is a currently untreatable form of melanoma with a 50% mortality rate. Characterization of the essential signaling pathways driving this cancer is critical to develop target therapies. Activating mutations in the Gαq signaling pathway at the level of GNAQ, GNA11, or rarely CYSLTR2 or PLCβ4 are considered alterations driving proliferation in UM and several other neoplastic disorders. Here, we systematically examined the oncogenic signaling output of various mutations recurrently identified in human tumors. We demonstrate that CYSLTR2 → GNAQ/11 → PLCβ act in a linear signaling cascade that, via protein kinase C (PKC), activates in parallel the MAP-kinase and FAK/Yes-associated protein pathways. Using genetic ablation and pharmacological inhibition, we show that the PKC/RasGRP3/MAPK signaling branch is the essential component that drives the proliferation of UM. Only inhibition of the MAPK branch but not the FAK branch synergizes with inhibition of the proximal cascade, providing a blueprint for combination therapy. All oncogenic signaling could be extinguished by the novel GNAQ/11 inhibitor YM-254890, in all UM cells with driver mutation in the Gαq subunit or the upstream receptor. Our findings highlight the GNAQ/11 → PLCβ → PKC → MAPK pathway as the central signaling axis to be suppressed pharmacologically to treat for neoplastic disorders with Gαq pathway mutations.
Collapse
|
45
|
Gaffal E. Forschung in der Praxis: Onkogene
GNAQ
‐Mutationen als therapeutischer Angriffspunkt beim Uveamelanom. J Dtsch Dermatol Ges 2020; 18:1245-1249. [DOI: 10.1111/ddg.14288_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/21/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Evelyn Gaffal
- Universitätshautklinik Universitätsklinikum Magdeburg
| |
Collapse
|
46
|
Katan M, Cockcroft S. Phospholipase C families: Common themes and versatility in physiology and pathology. Prog Lipid Res 2020; 80:101065. [PMID: 32966869 DOI: 10.1016/j.plipres.2020.101065] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/14/2020] [Accepted: 09/17/2020] [Indexed: 12/20/2022]
Abstract
Phosphoinositide-specific phospholipase Cs (PLCs) are expressed in all mammalian cells and play critical roles in signal transduction. To obtain a comprehensive understanding of these enzymes in physiology and pathology, a detailed structural, biochemical, cell biological and genetic information is required. In this review, we cover all these aspects to summarize current knowledge of the entire superfamily. The families of PLCs have expanded from 13 enzymes to 16 with the identification of the atypical PLCs in the human genome. Recent structural insights highlight the common themes that cover not only the substrate catalysis but also the mechanisms of activation. This involves the release of autoinhibitory interactions that, in the absence of stimulation, maintain classical PLC enzymes in their inactive forms. Studies of individual PLCs provide a rich repertoire of PLC function in different physiologies. Furthermore, the genetic studies discovered numerous mutated and rare variants of PLC enzymes and their link to human disease development, greatly expanding our understanding of their roles in diverse pathologies. Notably, substantial evidence now supports involvement of different PLC isoforms in the development of specific cancer types, immune disorders and neurodegeneration. These advances will stimulate the generation of new drugs that target PLC enzymes, and will therefore open new possibilities for treatment of a number of diseases where current therapies remain ineffective.
Collapse
Affiliation(s)
- Matilda Katan
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| | - Shamshad Cockcroft
- Department of Neuroscience, Physiology and Pharmacology, Division of Biosciences, University College London, 21 University Street, London WC1E 6JJ, UK.
| |
Collapse
|
47
|
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: 35] [Impact Index Per Article: 7.0] [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
|
48
|
Mallone F, Sacchetti M, Lambiase A, Moramarco A. Molecular Insights and Emerging Strategies for Treatment of Metastatic Uveal Melanoma. Cancers (Basel) 2020; 12:E2761. [PMID: 32992823 PMCID: PMC7600598 DOI: 10.3390/cancers12102761] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022] Open
Abstract
Uveal melanoma (UM) is the most common intraocular cancer. In recent decades, major advances have been achieved in the diagnosis and prognosis of UM allowing for tailored treatments. However, nearly 50% of patients still develop metastatic disease with survival rates of less than 1 year. There is currently no standard of adjuvant and metastatic treatment in UM, and available therapies are ineffective resulting from cutaneous melanoma protocols. Advances and novel treatment options including liver-directed therapies, immunotherapy, and targeted-therapy have been investigated in UM-dedicated clinical trials on single compounds or combinational therapies, with promising results. Therapies aimed at prolonging or targeting metastatic tumor dormancy provided encouraging results in other cancers, and need to be explored in UM. In this review, the latest progress in the diagnosis, prognosis, and treatment of UM in adjuvant and metastatic settings are discussed. In addition, novel insights into tumor genetics, biology and immunology, and the mechanisms underlying metastatic dormancy are discussed. As evident from the numerous studies discussed in this review, the increasing knowledge of this disease and the promising results from testing of novel individualized therapies could offer future perspectives for translating in clinical use.
Collapse
Affiliation(s)
| | | | - Alessandro Lambiase
- Department of Sense Organs, Sapienza University of Rome, 00161 Rome, Italy; (F.M.); (M.S.); (A.M.)
| | | |
Collapse
|
49
|
Kuschak M, Schlegel JG, Schneider M, Kehraus S, Voss JH, Seidinger A, Matthey M, Wenzel D, Fleischmann BK, König GM, Müller CE. Sensitive LC-MS/MS Method for the Quantification of Macrocyclic Gα q Protein Inhibitors in Biological Samples. Front Chem 2020; 8:833. [PMID: 33173765 PMCID: PMC7540253 DOI: 10.3389/fchem.2020.00833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/07/2020] [Indexed: 12/26/2022] Open
Abstract
The cyclic depsipeptide FR900359 (FR) isolated from the plant Ardisia crenata and produced by endosymbiotic bacteria acts as a selective Gq protein inhibitor. It is a powerful tool to study G protein-coupled receptor signaling, and has potential as a novel drug for the treatment of pulmonary diseases and cancer. For pharmacokinetic studies, sensitive quantitative measurements of drug levels are required. In the present study we established an LC-MS/MS method to detect nanomolar concentrations of FR and the structurally related natural product YM-254890 (YM) in biological samples. HPLC separation coupled to ESI-QTOF-MS and UV-VIS detection was applied. For identification and quantification, the extract ion chromatogram (EIC) of M+1 was evaluated. Limits of detection (LOD) of 0.53-0.55 nM and limits of quantification (LOQ) of 1.6-1.7 nM were achieved for both FR and YM. This protocol was subsequently applied to determine FR concentrations in mouse organs and tissues after peroral application of the drug. A three-step liquid-liquid extraction protocol was established, which resulted in adequate recovery rates of typically around 50%. The results indicated low peroral absorption of FR. Besides the gut, highest concentrations were determined in eye and kidney. The developed analytical method will be useful for preclinical studies to evaluate these potent Gq protein inhibitors, which may have potential as future drugs for complex diseases.
Collapse
Affiliation(s)
- Markus Kuschak
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Jonathan G Schlegel
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Marion Schneider
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Stefan Kehraus
- Institute of Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Jan H Voss
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Alexander Seidinger
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Bonn, Germany.,Department of Systems Physiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Michaela Matthey
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Bonn, Germany.,Department of Systems Physiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Daniela Wenzel
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Bonn, Germany.,Department of Systems Physiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Bernd K Fleischmann
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Bonn, Germany
| | - Gabriele M König
- Institute of Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn, Germany
| |
Collapse
|
50
|
Gaffal E. Research in practice: Therapeutic targeting of oncogenic GNAQ mutations in uveal melanoma. J Dtsch Dermatol Ges 2020; 18:1245-1248. [PMID: 32954611 DOI: 10.1111/ddg.14288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/21/2020] [Indexed: 02/03/2023]
Abstract
Uveal melanoma is the most common form of eye cancer and has a poor prognosis. Although the primary tumor in most cases is treated effectively by local surgery or radiotherapy, over 50 % of patients develop systemic metastasis, especially in the liver. In contrast to cutaneous melanoma, there is no standard-of-care treatment for metastasized uveal melanoma. Recently, oncogenic driver mutations in GNAQ or GNA11 were identified in about 85 % of uveal melanomas, which lead to constitutively active signaling in the Gαq/11 pathway and its downstream effectors. Direct targeting of deregulated Gαq/11 signaling might therefore be a therapeutic option for patients with uveal melanoma. In our study we identified the cyclic depsipeptide FR-900359, which is isolated from the evergreen plant Ardisia crenata as an effective inhibitor of constitutively active Gαq/11 proteins and their downstream targets. Although our data are preliminary, they might contribute to a future treatment option for patients with metastasized uveal melanoma.
Collapse
Affiliation(s)
- Evelyn Gaffal
- Department of Dermatology, University Hospital of Magdeburg, Magdeburg, Germany
| |
Collapse
|