1
|
Tao R, Li Y, Gong S, Zhang Q, Zhu Z. Unveiling intricating roles and mechanisms of ferroptosis in melanoma. Biochim Biophys Acta Rev Cancer 2024; 1880:189234. [PMID: 39644939 DOI: 10.1016/j.bbcan.2024.189234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
Melanoma is a highly invasive malignant tumor originating from melanocytes, with increasing incidence in recent years. Ferroptosis is an iron-dependent and non-apoptotic form of programmed cell death characterized by the accumulation of lipid peroxides and reactive oxygen species. Morphologically, ferroptosis exhibits the alteration in cells, such as reduced mitochondrial volume, increased density of bilayer membrane, and a decrease or disappearance of mitochondrial cristae. Ferroptosis has shown tremendous potential and applicability in regulating the development of melanoma. As melanoma progresses, certain biomarkers associated with ferroptosis display characteristic patterns of expression. These changes not only reveal the sensitivity of tumor cells to ferroptosis but also provide potential targets for diagnosis and treatment. Besides, inducing ferroptosis has been well-documented to inhibit the growth of melanoma and enhance the efficacy of tumor immunotherapy. Hence, this review emphasizes the roles and regulatory mechanisms of ferroptosis in melanoma development, the involved immune regulation, as well as the potential for diagnosis and treatment of melanoma. The continuous explorations will endow novel strategies for developing ferroptosis-based therapies for melanoma.
Collapse
Affiliation(s)
- Rui Tao
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Yichuan Li
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Song Gong
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China; Xianning Medical College, Hubei University of Science & Technology, Xianning 437000, Hubei Province, China.
| | - Zhanyong Zhu
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China.
| |
Collapse
|
2
|
Tsimpidakis A, Koumprentziotis IA, Mastoraki E, Plaka M, Gogas H, Stratigos A, Nikolaou V. Exacerbation of Kaposi sarcoma following BRAF/MEK inhibitor therapy in a melanoma patient: a case report and mechanistic insight. Melanoma Res 2024; 34:469-471. [PMID: 39037715 DOI: 10.1097/cmr.0000000000000991] [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: 07/23/2024]
Abstract
We present a case of a 75-year-old male patient who experienced a severe exacerbation of his Kaposi sarcoma lesions, which have remained clinically stable for a year, following treatment with BRAF/mitogen-activated protein kinase inhibitors for his coexisting melanoma. In this case, we present the possibility that BRAF/MEK inhibition may be mechanistically associated with the progression of Kaposi sarcoma and briefly discuss the potential mechanisms behind this phenomenon.
Collapse
Affiliation(s)
- Antonios Tsimpidakis
- First Department of Dermatology-Venereology, Medical School, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases
| | - Ioannis-Alexios Koumprentziotis
- First Department of Dermatology-Venereology, Medical School, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases
| | - Evanthia Mastoraki
- First Department of Dermatology-Venereology, Medical School, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases
| | - Michaella Plaka
- First Department of Dermatology-Venereology, Medical School, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases
| | - Helen Gogas
- First Department of Medicine, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexander Stratigos
- First Department of Dermatology-Venereology, Medical School, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases
| | - Vasiliki Nikolaou
- First Department of Dermatology-Venereology, Medical School, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases
| |
Collapse
|
3
|
Yao L, Krasnick BA, Bi Y, Sethuraman S, Goedegebuure S, Weerasinghe A, Wetzel C, Gao Q, Oyedeji A, Mudd J, Wyczalkowski MA, Wendl M, Ding L, Fields RC. Treatment resistance to melanoma therapeutics on a single cell level. Sci Rep 2024; 14:21915. [PMID: 39300183 DOI: 10.1038/s41598-024-72255-9] [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: 01/28/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024] Open
Abstract
Therapy targeting the BRAF-MEK cascade created a treatment revolution for patients with BRAF mutant advanced melanoma. Unfortunately, 80% patients treated will progress by 5 years follow-up. Thus, it is imperative we study mechanisms of melanoma progression and therapeutic resistance. We created a scRNA (single cell RNA) atlas of 128,230 cells from 18 tumors across the treatment spectrum, discovering melanoma cells clustered strongly by transcriptome profiles of patients of origins. Our cell-level investigation revealed gains of 1q and 7q as likely early clonal events in metastatic melanomas. By comparing patient tumors and their derivative cell lines, we observed that PD1 responsive tumor fraction disappears when cells are propagated in vitro. We further established three anti-BRAF-MEK treatment resistant cell lines using three BRAF mutant tumors. ALDOA and PGK1 were found to be highly expressed in treatment resistant cell populations and metformin was effective in targeting the resistant cells. Our study suggests that the investigation of patient tumors and their derivative lines is essential for understanding disease progression, treatment response and resistance.
Collapse
Affiliation(s)
- Lijun Yao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Bradley A Krasnick
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Ye Bi
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Sunantha Sethuraman
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Simon Goedegebuure
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Amila Weerasinghe
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Chris Wetzel
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Qingsong Gao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Abimbola Oyedeji
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Jacqueline Mudd
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Michael Wendl
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA.
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| | - Ryan C Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA.
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| |
Collapse
|
4
|
Volpe-Fix AR, da Silva Sales F, Caperuto LC, Lago JHG, Machado-Jr J. Essential Oil Extracted from Lippia sidoides Cham. (Verbenaceae) Induces Cell Cycle Arrest, Apoptosis, and Antimigratory Effects in Melanoma Cells. Chem Biodivers 2024:e202401530. [PMID: 39269919 DOI: 10.1002/cbdv.202401530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 09/15/2024]
Abstract
Melanoma, the most aggressive form of skin cancer, poses a substantial global health threat with increasing incidence rates. Although novel targeted therapies have improved melanoma treatment, challenges persist due to poor response rates and drug resistance. Plant-derived compounds have been crucial in anticancer drug discovery, with many natural products demonstrating the ability to target molecular pathways involved in tumor development. In this study, the anti-melanoma potential of essential oil extracted from the aerial parts of Lippia sidoides Cham. (EO-LS), composed mainly by the monoterpene thymol (96 %), was demonstrated. Obtained results demonstrated that EO-LS disrupted critical cancer hallmarks in A2058 melanoma cells harboring the BRAFV600E mutation. Specifically, EO-LS induced G1-phase cell cycle arrest and apoptosis, as assessed by annexin-V, caspase-3 activity, and TUNEL assays. EO-LS also inhibited cell migration and disrupted the AKT signaling pathway, which is a critical regulator of melanoma progression. Furthermore, a dose-dependent increase in reactive oxygen species (ROS) generation was observed, indicating pro-oxidant properties. These findings highlighted the significant in vitro anticancer properties of EO-LS suggesting its potential as a promising molecular scaffold for developing of novel anti-melanoma candidates.
Collapse
Affiliation(s)
- Andressa Roehrig Volpe-Fix
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, 09972-270, Brazil
| | - Felipe da Silva Sales
- Center for Natural and Human Sciences, Federal University of ABC, São Paulo, 092210-180, SP Brazil
| | - Luciana Chagas Caperuto
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, 09972-270, Brazil
| | | | - Joel Machado-Jr
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, 09972-270, Brazil
| |
Collapse
|
5
|
Kot M, Simiczyjew A, Wądzyńska J, Ziętek M, Matkowski R, Nowak D. Characterization of two melanoma cell lines resistant to BRAF/MEK inhibitors (vemurafenib and cobimetinib). Cell Commun Signal 2024; 22:410. [PMID: 39175042 PMCID: PMC11342534 DOI: 10.1186/s12964-024-01788-3] [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: 02/22/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND BRAF (v-raf murine sarcoma viral oncogene homolog B1)/MEK (mitogen-activated protein kinase kinase) inhibitors are used for melanoma treatment. Unfortunately, patients treated with this combined therapy develop resistance to treatment quite quickly, but the mechanisms underlying this phenomenon are not yet fully understood. Here, we report and characterize two melanoma cell lines (WM9 and Hs294T) resistant to BRAF (vemurafenib) and MEK (cobimetinib) inhibitors. METHODS Cell viability was assessed via the XTT test. The level of selected proteins as well as activation of signaling pathways were evaluated using Western blotting. The expression of the chosen genes was assessed by RT-PCR. The distribution of cell cycle phases was analyzed by flow cytometry, and confocal microscopy was used to take photos of spheroids. The composition of cytokines secreted by cells was determined using a human cytokine array. RESULTS The resistant cells had increased survival and activation of ERK kinase in the presence of BRAF/MEK inhibitors. The IC50 values for these cells were over 1000 times higher than for controls. Resistant cells also exhibited elevated activation of AKT, p38, and JNK signaling pathways with increased expression of EGFR, ErbB2, MET, and PDGFRβ receptors as well as reduced expression of ErbB3 receptor. Furthermore, these cells demonstrated increased expression of genes encoding proteins involved in drug transport and metabolism. Resistant cells also exhibited features of epithelial-mesenchymal transition and cancer stem cells as well as reduced proliferation rate and elevated cytokine secretion. CONCLUSIONS In summary, this work describes BRAF/MEK-inhibitor-resistant melanoma cells, allowing for better understanding the underlying mechanisms of resistance. The results may thus contribute to the development of new, more effective therapeutic strategies.
Collapse
Affiliation(s)
- Magdalena Kot
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Aleksandra Simiczyjew
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland.
| | - Justyna Wądzyńska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Marcin Ziętek
- Department of Oncology, Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
- Lower Silesian Oncology, Pulmonology, and Hematology Center, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
| | - Rafał Matkowski
- Department of Oncology, Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
- Lower Silesian Oncology, Pulmonology, and Hematology Center, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| |
Collapse
|
6
|
Gu R, Fang H, Wang R, Dai W, Cai G. A comprehensive overview of the molecular features and therapeutic targets in BRAF V600E-mutant colorectal cancer. Clin Transl Med 2024; 14:e1764. [PMID: 39073010 DOI: 10.1002/ctm2.1764] [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: 05/07/2024] [Revised: 06/23/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
As one of the most prevalent digestive system tumours, colorectal cancer (CRC) poses a significant threat to global human health. With the emergence of immunotherapy and target therapy, the prognosis for the majority of CRC patients has notably improved. However, the subset of patients with BRAF exon 15 p.V600E mutation (BRAFV600E) has not experienced remarkable benefits from these therapeutic advancements. Hence, researchers have undertaken foundational investigations into the molecular pathology of this specific subtype and clinical effectiveness of diverse therapeutic drug combinations. This review comprehensively summarised the distinctive molecular features and recent clinical research advancements in BRAF-mutant CRC. To explore potential therapeutic targets, this article conducted a systematic review of ongoing clinical trials involving patients with BRAFV600E-mutant CRC.
Collapse
Affiliation(s)
- Ruiqi Gu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongsheng Fang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Renjie Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weixing Dai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guoxiang Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Németh A, Bányai GL, Dobos NK, Kós T, Gaál A, Varga Z, Buzás EI, Khamari D, Dank M, Takács I, Szász AM, Garay T. Extracellular vesicles promote migration despite BRAF inhibitor treatment in malignant melanoma cells. Cell Commun Signal 2024; 22:282. [PMID: 38778340 PMCID: PMC11110207 DOI: 10.1186/s12964-024-01660-4] [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: 08/14/2023] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Extracellular vesicles (EVs) constitute a vital component of intercellular communication, exerting significant influence on metastasis formation and drug resistance mechanisms. Malignant melanoma (MM) is one of the deadliest forms of skin cancers, because of its high metastatic potential and often acquired resistance to oncotherapies. The prevalence of BRAF mutations in MM underscores the importance of BRAF-targeted therapies, such as vemurafenib and dabrafenib, alone or in combination with the MEK inhibitor, trametinib. This study aimed to elucidate the involvement of EVs in MM progression and ascertain whether EV-mediated metastasis promotion persists during single agent BRAF (vemurafenib, dabrafenib), or MEK (trametinib) and combined BRAF/MEK (dabrafenib/trametinib) inhibition.Using five pairs of syngeneic melanoma cell lines, we assessed the impact of EVs - isolated from their respective supernatants - on melanoma cell proliferation and migration. Cell viability and spheroid growth assays were employed to evaluate proliferation, while migration was analyzed through mean squared displacement (MSD) and total traveled distance (TTD) measurements derived from video microscopy and single-cell tracking.Our results indicate that while EV treatments had remarkable promoting effect on cell migration, they exerted only a modest effect on cell proliferation and spheroid growth. Notably, EVs demonstrated the ability to mitigate the inhibitory effects of BRAF inhibitors, albeit they were ineffective against a MEK inhibitor and the combination of BRAF/MEK inhibitors. In summary, our findings contribute to the understanding of the intricate role played by EVs in tumor progression, metastasis, and drug resistance in MM.
Collapse
Affiliation(s)
- Afrodité Németh
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Gréta L Bányai
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Nikolett K Dobos
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Tamás Kós
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Anikó Gaál
- Institute of Materials and Environmental Chemistry; Biological Nanochemistry Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Zoltán Varga
- Institute of Materials and Environmental Chemistry; Biological Nanochemistry Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- ELKH-SE Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HCEMM-SE Extracellular Vesicle Research Group, Budapest, Hungary
| | - Delaram Khamari
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Magdolna Dank
- Department of Internal Medicine and Oncology, Division of Oncology, Semmelweis University, Budapest, Hungary
| | - István Takács
- Department of Internal Medicine and Oncology, Division of Oncology, Semmelweis University, Budapest, Hungary
| | - A Marcell Szász
- Department of Internal Medicine and Oncology, Division of Oncology, Semmelweis University, Budapest, Hungary
| | - Tamás Garay
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary.
- Department of Internal Medicine and Oncology, Division of Oncology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
8
|
Yan C, Zhao L, Zhang X, Chu Z, Zhou T, Zhang Y, Geng S, Guo K. Cold atmospheric plasma sensitizes melanoma cells to targeted therapy agents in vitro. JOURNAL OF BIOPHOTONICS 2024; 17:e202300356. [PMID: 38041219 DOI: 10.1002/jbio.202300356] [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: 09/03/2023] [Revised: 11/15/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023]
Abstract
Cold atmospheric plasma (CAP) has been reported to kill melanoma cells in vitro and in vivo. BRAF and MEK inhibitors are targeted therapy agents for advanced melanoma patients with BRAF mutations. However, low overall survival and relapse-free survival are still tough challenges due to drug resistance. In this study, we confirmed that CAP alleviated innate drug resistance and promoted the anti-tumor effect of targeted therapy in A875 and WM115 melanoma cells in vitro. Further, we revealed that CAP altered the expression of various molecules concerning MAPK and PI3K-AKT pathways in A875 cells. This study demonstrates that CAP promises to work as adjuvant treatment with targeted therapy to overcome drug resistance for malignant tumors in future.
Collapse
Affiliation(s)
- Cong Yan
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lihong Zhao
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xinyue Zhang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhaowei Chu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Tong Zhou
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yanbin Zhang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Songmei Geng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Center for Dermatology Disease, Precision Medical Institute, Xi'an, China
| | - Kun Guo
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Center for Dermatology Disease, Precision Medical Institute, Xi'an, China
| |
Collapse
|
9
|
Cammann C, Kulla J, Wiebusch L, Walz C, Zhao F, Lowinus T, Topfstedt E, Mishra N, Henklein P, Bommhardt U, Bossaller L, Hagemeier C, Schadendorf D, Schmidt B, Paschen A, Seifert U. Proteasome inhibition potentiates Kv1.3 potassium channel expression as therapeutic target in drug-sensitive and -resistant human melanoma cells. Biomed Pharmacother 2023; 168:115635. [PMID: 37816303 DOI: 10.1016/j.biopha.2023.115635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/24/2023] [Accepted: 10/03/2023] [Indexed: 10/12/2023] Open
Abstract
Primary and acquired therapy resistance is a major problem in patients with BRAF-mutant melanomas being treated with BRAF and MEK inhibitors (BRAFI, MEKi). Therefore, development of alternative therapy regimes is still required. In this regard, new drug combinations targeting different pathways to induce apoptosis could offer promising alternative approaches. Here, we investigated the combination of proteasome and Kv1.3 potassium channel inhibition on chemo-resistant, BRAF inhibitor-resistant as well as sensitive human melanoma cells. Our experiments demonstrated that all analyzed melanoma cell lines were sensitive to proteasome inhibitor treatment at concentrations that are not toxic to primary human fibroblasts. To further reduce proteasome inhibitor-associated side effects, and to foster apoptosis, potassium channels, which are other targets to induce pro-apoptotic effects in cancer cells, were blocked. In support, combined exposure of melanoma cells to proteasome and Kv1.3 channel inhibitor resulted in synergistic effects and significantly reduced cell viability. On the molecular level, enhanced apoptosis correlated with an increase of intracellular Kv1.3 channels and pro-apoptotic proteins such as Noxa and Bak and a reduction of anti-apoptotic proteins. Thus, use of combined therapeutic strategies triggering different apoptotic pathways may efficiently prevent the outgrowth of drug-resistant and -sensitive BRAF-mutant melanoma cells. In addition, this could be the basis for an alternative approach to treat other tumors expressing mutated BRAF such as non-small-cell lung cancer.
Collapse
Affiliation(s)
- Clemens Cammann
- Friedrich Loeffler - Institute of Medical Microbiology - Virology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Jonas Kulla
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke-University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Lüder Wiebusch
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Christian Walz
- Clemens Schöpf-Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Alarich Weiss-Straße 4-8, 64287 Darmstadt, Germany
| | - Fang Zhao
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Theresa Lowinus
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke-University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Eylin Topfstedt
- Friedrich Loeffler - Institute of Medical Microbiology - Virology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Neha Mishra
- Section of Rheumatology, Clinic and Policlinic of Internal Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Petra Henklein
- Institute of Molecular Biology and Biochemistry, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Ursula Bommhardt
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke-University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Lukas Bossaller
- Section of Rheumatology, Clinic and Policlinic of Internal Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Christian Hagemeier
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Boris Schmidt
- Clemens Schöpf-Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Alarich Weiss-Straße 4-8, 64287 Darmstadt, Germany
| | - Annette Paschen
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Ulrike Seifert
- Friedrich Loeffler - Institute of Medical Microbiology - Virology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany.
| |
Collapse
|
10
|
Batsi Y, Antonopoulou G, Fotopoulou T, Koumaki K, Kritsi E, Potamitis C, Goulielmaki M, Skarmalioraki S, Papalouka C, Poulou-Sidiropoulou E, Kosmidou V, Douna S, Vidali MS, Gkotsi EF, Chatziioannou A, Souliotis VL, Pletsa V, Papadodima O, Zoumpourlis V, Georgiadis P, Zervou M, Pintzas A, Kostas ID. Design and Synthesis of Novel 2-Acetamido, 6-Carboxamide Substituted Benzothiazoles as Potential BRAFV600E Inhibitors - In vitro Evaluation of their Antiproliferative Activity. ChemMedChem 2023; 18:e202300322. [PMID: 37792577 DOI: 10.1002/cmdc.202300322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/06/2023]
Abstract
The oncogenic BRAFV600E kinase leads to abnormal activation of the MAPK signaling pathway and thus, uncontrolled cellular proliferation and cancer development. Based on our previous virtual screening studies which issued 2-acetamido-1,3 benzothiazole-6-carboxamide scaffold as active pharmacophore displaying selectivity against the mutated BRAF, eleven new substituted benzothiazole derivatives were designed and synthesized by coupling of 2-acetamidobenzo[d]thiazole-6-carboxylic acid with the appropriate amines in an effort to provide even more efficient inhibitors and tackle drug resistance often developed during cancer treatment. All derived compounds bore the benzothiazole scaffold substituted at position-2 by an acetamido moiety and at position-6 by a carboxamide functionality, the NH moiety of which was further linked through an alkylene linker to a sulfonamido (or amino) aryl (or alkyl) functionality or a phenylene linker to a sulfonamido aromatic (or non-aromatic) terminal pharmacophore in the order -C6 H4 -NHSO2 -R or reversely -C6 H4 -SO2 N(H)-R. These analogs were subsequently biologically evaluated as potential BRAFV600E inhibitors and antiproliferative agents in several colorectal cancer and melanoma cell lines. In all assays applied, one analog, namely 2-acetamido-N-[3-(pyridin-2-ylamino)propyl]benzo[d]thiazole-6-carboxamide (22), provided promising results in view of its use in drug development.
Collapse
Affiliation(s)
- Yakinthi Batsi
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Georgia Antonopoulou
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Theano Fotopoulou
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Kassandra Koumaki
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Eftichia Kritsi
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Constantinos Potamitis
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Maria Goulielmaki
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Salomi Skarmalioraki
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Chara Papalouka
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Eleni Poulou-Sidiropoulou
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Vivian Kosmidou
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Stavroula Douna
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Maria-Sofia Vidali
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Eleni-Fani Gkotsi
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Aristotelis Chatziioannou
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Vassilis L Souliotis
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Vasiliki Pletsa
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Olga Papadodima
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Vassilis Zoumpourlis
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Panagiotis Georgiadis
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Maria Zervou
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Alexander Pintzas
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| | - Ioannis D Kostas
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou Ave. 48, 11635, Athens, Greece
| |
Collapse
|
11
|
Gambichler T, Harnischfeger F, Skrygan M, Majchrzak-Stiller B, Buchholz M, Müller T, Braumann C. In Vitro Experiments on the Effects of GP-2250 on BRAF-Mutated Melanoma Cell Lines and Benign Melanocytes. Int J Mol Sci 2023; 24:15336. [PMID: 37895015 PMCID: PMC10607550 DOI: 10.3390/ijms242015336] [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: 09/05/2023] [Revised: 10/15/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Enhanced glycolysis (Warburg effect) driven by the BRAF oncogene, dysregulated GAPDH expression, and activation of the PI3K/AKT/mTOR signaling pathway may significantly contribute to the resistance-targeted therapy of BRAF-mutated melanomas. Therefore, we aimed to study for the first time the anti-tumor activity of the GAPDH inhibitor GP-2250 in BRAF-mutated melanoma cell lines and benign melanocytes. We employed three melanoma cell lines and one primary melanocyte cell line (Ma-Mel-61a, Ma-Mel-86a, SH-4 and ATCC-PCS-200-013, respectively), which were exposed to different GP-2250 doses. GP-2250's effects on cell proliferation and viability were evaluated by means of the BrdU and MTT assays, respectively. The RealTime-Glo Annexin V Apoptosis and Necrosis Assay was performed for the evaluation of apoptosis and necrosis induction. RT-PCR and western blotting were implemented for the determination of AKT and STAT3 gene and protein expression analyses, respectively. The melanoma cell lines showed a dose-dependent response to GP-2250 during BrDU and MTT testing. The RealTime-Glo Annexin V assay revealed the heterogenous impact of GP-2250 on apoptosis as well as necrosis. With respect to the melanoma cell lines Ma-Mel-86a and SH-4, the responses and dosages were comparable to those used for the MTT viability assay. Using the same dose range of GP-2250 administered to melanoma cells, however, we observed neither the noteworthy apoptosis nor necrosis of GP-2250-treated benign melanocytes. The gene expression profiles in the melanoma cell lines for AKT and STAT3 were heterogenous, whereby AKT as well as STAT3 gene expression were most effectively downregulated using the highest GP-2250 doses. Immunoblotting revealed that there was a time-dependent decrease in protein expression at the highest GP-2250 dose used, whereas a time- as well as dose-dependent AKT decrease was predominantly observed in Ma-Mel-61a. The STAT3 protein expression of Ma-Mel-86a and SH-4 was reduced in a time-dependent pattern at lower and moderate doses. STAT3 expression in Ma-Me-61a was barely altered by GP-2250. In conclusion, GP-2250 has anti-neoplastic effects in BRAF-mutated melanoma cell lines regarding tumor cell viability, proliferation, and apoptosis/necrosis. GP-2250 is able to downregulate the gene and protein expression of aberrant tumorigenic pathways in melanoma cell lines. Since GP-2250 is a GAPDH inhibitor, the substance may be a promising combination therapy for tumors presenting the Warburg effect, such as melanoma.
Collapse
Affiliation(s)
- Thilo Gambichler
- Skin Cancer Center Ruhr-University, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany; (F.H.); (M.S.)
| | - Friederike Harnischfeger
- Skin Cancer Center Ruhr-University, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany; (F.H.); (M.S.)
| | - Marina Skrygan
- Skin Cancer Center Ruhr-University, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany; (F.H.); (M.S.)
| | - Britta Majchrzak-Stiller
- Department of General and Visceral Surgery, Division of Molecular and Clinical Research, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany; (B.M.-S.); (M.B.)
| | - Marie Buchholz
- Department of General and Visceral Surgery, Division of Molecular and Clinical Research, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany; (B.M.-S.); (M.B.)
| | | | - Chris Braumann
- Department of General, Visceral and Vascular Surgery, Evangelisches Klinikum Gelsenkirchen, Akademisches Lehrkrankenhaus der Universität Duisburg-Essen, 45878 Gelsenkirchen, Germany;
- Department of General, Visceral and Tumor Surgery, Evangelisches Klinikum Herne, Akademisches Lehrkrankenhaus der Ruhr-Universität Bochum, 44623 Herne, Germany
| |
Collapse
|
12
|
Castellani G, Buccarelli M, Arasi MB, Rossi S, Pisanu ME, Bellenghi M, Lintas C, Tabolacci C. BRAF Mutations in Melanoma: Biological Aspects, Therapeutic Implications, and Circulating Biomarkers. Cancers (Basel) 2023; 15:4026. [PMID: 37627054 PMCID: PMC10452867 DOI: 10.3390/cancers15164026] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Melanoma is an aggressive form of skin cancer resulting from the malignant transformation of melanocytes. Recent therapeutic approaches, including targeted therapy and immunotherapy, have improved the prognosis and outcome of melanoma patients. BRAF is one of the most frequently mutated oncogenes recognised in melanoma. The most frequent oncogenic BRAF mutations consist of a single point mutation at codon 600 (mostly V600E) that leads to constitutive activation of the BRAF/MEK/ERK (MAPK) signalling pathway. Therefore, mutated BRAF has become a useful target for molecular therapy and the use of BRAF kinase inhibitors has shown promising results. However, several resistance mechanisms invariably develop leading to therapeutic failure. The aim of this manuscript is to review the role of BRAF mutational status in the pathogenesis of melanoma and its impact on differentiation and inflammation. Moreover, this review focuses on the mechanisms responsible for resistance to targeted therapies in BRAF-mutated melanoma and provides an overview of circulating biomarkers including circulating tumour cells, circulating tumour DNA, and non-coding RNAs.
Collapse
Affiliation(s)
- Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Maria Beatrice Arasi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Stefania Rossi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Maria Elena Pisanu
- High Resolution NMR Unit, Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Maria Bellenghi
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Carla Lintas
- Research Unit of Medical Genetics, Department of Medicine, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
- Operative Research Unit of Medical Genetics, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Claudio Tabolacci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| |
Collapse
|
13
|
Huang X, Gou W, Song Q, Huang Y, Wen C, Bo X, Jiang X, Feng J, Gao H. A BRAF mutation-associated gene risk model for predicting the prognosis of melanoma. Heliyon 2023; 9:e15939. [PMID: 37205993 PMCID: PMC10189240 DOI: 10.1016/j.heliyon.2023.e15939] [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: 02/02/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/21/2023] Open
Abstract
BRAF mutation plays an important role in the pathogenesis and progression of melanoma and is correlated to the prognosis of melanoma patients. However, fewer studies have attempted to develop a BRAF mutation-associated gene risk model for predicting the prognosis of melanoma. The current research explores BRAF mutation-related biological features in melanoma and establishes a prognostic signature. First, we identified three significantly enriched KEGG pathways (glycosphingolipid biosynthesis - ganglio series, ether lipid metabolism, and glycosaminoglycan biosynthesis - keratan sulfate) and corresponding genes in the BRAF mutant group by gene set enrichment analysis. We then developed a prognostic signature based on 7 BRAF-associated genes (PLA2G2D, FUT8, PLA2G4E, PLA2G5, PLA2G1B, B3GNT2, and ST3GAL5) and assessed its prediction accuracy using ROC curve analysis. Finally, the nomogram was established according to the prognostic signature and independent clinical characteristics to predict the survival of melanoma patients. Furthermore, we found higher proportions of naive B cells, plasma cells, CD8 T cells, CD4 memory-activated T cells, and regulatory T cells in the low-risk group. Whereas lower proportions of M0, M1, and M2 macrophages and resting NK cells were observed in the high-risk group. The analysis also showed a significantly higher expression of immune checkpoint molecules (PD-1, PD-L1, CTLA4, BTLA, CD28, CD80, CD86, HAVCR2, ICOS, LAG3, and TIGIT) in the low-risk group. Our results provide novel insights into the effect of BRAF mutation on melanoma growth and indicate a promising direction toward immunotherapy and precision medicine in melanoma patients.
Collapse
Affiliation(s)
- Xiang Huang
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Wanrong Gou
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Qinxian Song
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yan Huang
- Department of Dermatology, Suining First People's Hospital, Suining, 629000, Sichuan, China
| | - Chunlei Wen
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Xue Bo
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Xian Jiang
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jianguo Feng
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Corresponding author.
| | - Hong Gao
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
- Corresponding author.
| |
Collapse
|
14
|
Vemurafenib and Dabrafenib Downregulates RIPK4 Level. Cancers (Basel) 2023; 15:cancers15030918. [PMID: 36765875 PMCID: PMC9913565 DOI: 10.3390/cancers15030918] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Vemurafenib and dabrafenib are BRAF kinase inhibitors (BRAFi) used for the treatment of patients with melanoma carrying the V600E BRAF mutation. However, melanoma cells develop resistance to both drugs when used as monotherapy. Therefore, mechanisms of drug resistance are investigated, and new molecular targets are sought that could completely inhibit melanoma progression. Since receptor-interacting protein kinase (RIPK4) probably functions as an oncogene in melanoma and its structure is similar to the BRAF protein, we analyzed the impact of vemurafenib and dabrafenib on RIPK4 in melanomas. The in silico study confirmed the high similarity of BRAF kinase domains to the RIPK4 protein at both the sequence and structural levels and suggests that BRAFi could directly bind to RIPK4 even more strongly than to ATP. Furthermore, BRAFi inhibited ERK1/2 activity and lowered RIPK4 protein levels in BRAF-mutated melanoma cells (A375 and WM266.4), while in wild-type BRAF cells (BLM and LoVo), both inhibitors decreased the level of RIPK4 and enhanced ERK1/2 activity. The phosphorylation of phosphatidylethanolamine binding protein 1 (PEBP1)-a suppressor of the BRAF/MEK/ERK pathway-via RIPK4 observed in pancreatic cancer did not occur in melanoma. Neither downregulation nor upregulation of RIPK4 in BRAF- mutated cells affected PEBP1 levels or the BRAF/MEK/ERK pathway. The downregulation of RIPK4 inhibited cell proliferation and the FAK/AKT pathway, and increased BRAFi efficiency in WM266.4 cells. However, the silencing of RIPK4 did not induce apoptosis or necroptosis. Our study suggests that RIPK4 may be an off-target for BRAF inhibitors.
Collapse
|
15
|
Sharma A, Sharma L, Nandy SK, Payal N, Yadav S, Vargas-De-La-Cruz C, Anwer MK, Khan H, Behl T, Bungau SG. Molecular Aspects and Therapeutic Implications of Herbal Compounds Targeting Different Types of Cancer. Molecules 2023; 28:750. [PMID: 36677808 PMCID: PMC9867434 DOI: 10.3390/molecules28020750] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/05/2023] [Accepted: 01/08/2023] [Indexed: 01/15/2023] Open
Abstract
Due to genetic changes in DNA (deoxyribonucleic acid) sequences, cancer continues to be the second most prevalent cause of death. The traditional target-directed approach, which is confronted with the importance of target function in healthy cells, is one of the most significant challenges in anticancer research. Another problem with cancer cells is that they experience various mutations, changes in gene duplication, and chromosomal abnormalities, all of which have a direct influence on the potency of anticancer drugs at different developmental stages. All of these factors combine to make cancer medication development difficult, with low clinical licensure success rates when compared to other therapy categories. The current review focuses on the pathophysiology and molecular aspects of common cancer types. Currently, the available chemotherapeutic drugs, also known as combination chemotherapy, are associated with numerous adverse effects, resulting in the search for herbal-based alternatives that attenuate resistance due to cancer therapy and exert chemo-protective actions. To provide new insights, this review updated the list of key compounds that may enhance the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Aditi Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, Himachal Pradesh, India
| | - Lalit Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, Himachal Pradesh, India
| | - Shouvik Kumar Nandy
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, Himachal Pradesh, India
| | - Nazrana Payal
- School of Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan 173229, Himachal Pradesh, India
| | - Shivam Yadav
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Chhatrapati Shahu ji Maharaj University, Kanpur 208024, Uttar Pradesh, India
| | - Celia Vargas-De-La-Cruz
- Department of Pharmacology, Faculty of Pharmacy and Biochemistry, Bromatology and Toxicology, Universidad Nacional Mayor de San Marcos, Lima 150001, Peru
- E-Health Research Center, Universidad de Ciencias y Humanidades, Lima 15001, Peru
| | - Md. Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Tapan Behl
- School of Health Science and Technology, University of Petroleum and Energy Studies, Dehradun 248007, Uttarakhand, India
| | - Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
- Doctoral School of Biomedical Sciences, University of Oradea, 410028 Oradea, Romania
| |
Collapse
|
16
|
Russi M, Valeri R, Marson D, Danielli C, Felluga F, Tintaru A, Skoko N, Aulic S, Laurini E, Pricl S. Some things old, new and borrowed: Delivery of dabrafenib and vemurafenib to melanoma cells via self-assembled nanomicelles based on an amphiphilic dendrimer. Eur J Pharm Sci 2023; 180:106311. [PMID: 36273785 DOI: 10.1016/j.ejps.2022.106311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Two clinically approved anticancer drugs targeting BRAF in melanoma patients - dabrafenib (DAB) and vemurafenib (VEM) - have been successfully encapsulated into nanomicelles formed upon self-assembly of an amphiphilic dendrimer AD based on two C18 aliphatic chains and a G2 PAMAM head. The process resulted in the formation of well-defined (∼10 nm) core-shell nanomicelles (NMs) with excellent encapsulation efficiency (∼70% for DAB and ∼60% for VEM) and good drug loading capacity (∼27% and ∼24% for DAB and VEM, respectively). Dynamic light scattering (DLS), transmission electron microscopy (TEM), small-angle x-ray scattering (SAXS), nuclear magnetic resonance (NMR), isothermal titration calorimetry (ITC), and molecular simulation (MS) experiments were used, respectively, to determine the size and structure of the empty and drug-loaded nanomicelles (DLNMs), along with the interactions between the NMs and their cargoes. The in vitro release data revealed profiles governed by Fickian diffusion; moreover, for both anticancer molecules, an acidic environment (pH = 5.0) facilitated drug release with respect to physiological pH conditions (pH = 7.4). Finally, both DAB- and VEM-loaded NMs elicited enhanced response with respect to free drug treatments in 4 different melanoma cell lines.
Collapse
Affiliation(s)
- Maria Russi
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS) - DEA, University of Trieste, Piazzale Europa 1, Trieste 34127, Italy
| | - Rachele Valeri
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS) - DEA, University of Trieste, Piazzale Europa 1, Trieste 34127, Italy
| | - Domenico Marson
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS) - DEA, University of Trieste, Piazzale Europa 1, Trieste 34127, Italy
| | - Chiara Danielli
- Department of Chemical and Pharmaceutical Sciences, DSCF, University of Trieste, Via Giorgeri 1, Trieste 34127, Italy
| | - Fulvia Felluga
- Department of Chemical and Pharmaceutical Sciences, DSCF, University of Trieste, Via Giorgeri 1, Trieste 34127, Italy
| | - Aura Tintaru
- Aix Marseille Univ, CNRS - Centre Interdisciplinaire de Nanosciences de Marseille (CINaM) UMR 7325 - Département IMMF - Campus Luminy, 163, Avenue de Luminy, Marseille 13288, France
| | - Natasa Skoko
- Biotechnology Development Unit, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Suzana Aulic
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS) - DEA, University of Trieste, Piazzale Europa 1, Trieste 34127, Italy; Biotechnology Development Unit, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Erik Laurini
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS) - DEA, University of Trieste, Piazzale Europa 1, Trieste 34127, Italy.
| | - Sabrina Pricl
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS) - DEA, University of Trieste, Piazzale Europa 1, Trieste 34127, Italy; Department of General Biophysics, University of Łódź, ul. Pomorska 141/143, Łódź 90-236, Poland
| |
Collapse
|
17
|
Identification of Dihydrolipoamide Dehydrogenase as Potential Target of Vemurafenib-Resistant Melanoma Cells. Molecules 2022; 27:molecules27227800. [PMID: 36431901 PMCID: PMC9698468 DOI: 10.3390/molecules27227800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Despite recent improvements in therapy, the five-year survival rate for patients with advanced melanoma is poor, mainly due to the development of drug resistance. The aim of the present study was to investigate the mechanisms underlying this phenomenon, applying proteomics and structural approaches to models of melanoma cells. METHODS Sublines from two human (A375 and SK-MEL-28) cells with acquired vemurafenib resistance were established, and their proteomic profiles when exposed to denaturation were identified through LC-MS/MS analysis. The pathways derived from bioinformatics analyses were validated by in silico and functional studies. RESULTS The proteomic profiles of resistant melanoma cells were compared to parental counterparts by taking into account protein folding/unfolding behaviors. Several proteins were found to be involved, with dihydrolipoamide dehydrogenase (DLD) being the only one similarly affected by denaturation in all resistant cell sublines compared to parental ones. DLD expression was observed to be increased in resistant cells by Western blot analysis. Protein modeling analyses of DLD's catalytic site coupled to in vitro assays with CPI-613, a specific DLD inhibitor, highlighted the role of DLD enzymatic functions in the molecular mechanisms of BRAFi resistance. CONCLUSIONS Our proteomic and structural investigations on resistant sublines indicate that DLD may represent a novel and potent target for overcoming vemurafenib resistance in melanoma cells.
Collapse
|
18
|
Houles T, Lavoie G, Nourreddine S, Cheung W, Vaillancourt-Jean É, Guérin CM, Bouttier M, Grondin B, Lin S, Saba-El-Leil MK, Angers S, Meloche S, Roux PP. CDK12 is hyperactivated and a synthetic-lethal target in BRAF-mutated melanoma. Nat Commun 2022; 13:6457. [PMID: 36309522 PMCID: PMC9617877 DOI: 10.1038/s41467-022-34179-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 10/13/2022] [Indexed: 12/25/2022] Open
Abstract
Melanoma is the deadliest form of skin cancer and considered intrinsically resistant to chemotherapy. Nearly all melanomas harbor mutations that activate the RAS/mitogen-activated protein kinase (MAPK) pathway, which contributes to drug resistance via poorly described mechanisms. Herein we show that the RAS/MAPK pathway regulates the activity of cyclin-dependent kinase 12 (CDK12), which is a transcriptional CDK required for genomic stability. We find that melanoma cells harbor constitutively high CDK12 activity, and that its inhibition decreases the expression of long genes containing multiple exons, including many genes involved in DNA repair. Conversely, our results show that CDK12 inhibition promotes the expression of short genes with few exons, including many growth-promoting genes regulated by the AP-1 and NF-κB transcription factors. Inhibition of these pathways strongly synergize with CDK12 inhibitors to suppress melanoma growth, suggesting promising drug combinations for more effective melanoma treatment.
Collapse
Affiliation(s)
- Thibault Houles
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, 2950, Chemin de la Polytechnique, Montréal, QC H3T 1J4 Canada
| | - Geneviève Lavoie
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, 2950, Chemin de la Polytechnique, Montréal, QC H3T 1J4 Canada
| | - Sami Nourreddine
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, 2950, Chemin de la Polytechnique, Montréal, QC H3T 1J4 Canada ,grid.266100.30000 0001 2107 4242Present Address: Department of Bioengineering, University of California, San Diego, San Diego, CA USA
| | - Winnie Cheung
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, 2950, Chemin de la Polytechnique, Montréal, QC H3T 1J4 Canada
| | - Éric Vaillancourt-Jean
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, 2950, Chemin de la Polytechnique, Montréal, QC H3T 1J4 Canada
| | - Célia M. Guérin
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, 2950, Chemin de la Polytechnique, Montréal, QC H3T 1J4 Canada
| | - Mathieu Bouttier
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, 2950, Chemin de la Polytechnique, Montréal, QC H3T 1J4 Canada
| | - Benoit Grondin
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, 2950, Chemin de la Polytechnique, Montréal, QC H3T 1J4 Canada ,grid.38678.320000 0001 2181 0211Present Address: Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC Canada
| | - Sichun Lin
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular & Biomolecular Research, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Marc K. Saba-El-Leil
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, 2950, Chemin de la Polytechnique, Montréal, QC H3T 1J4 Canada
| | - Stephane Angers
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular & Biomolecular Research, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Sylvain Meloche
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, 2950, Chemin de la Polytechnique, Montréal, QC H3T 1J4 Canada ,grid.14848.310000 0001 2292 3357Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC Canada
| | - Philippe P. Roux
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, 2950, Chemin de la Polytechnique, Montréal, QC H3T 1J4 Canada ,grid.14848.310000 0001 2292 3357Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montreal, QC Canada
| |
Collapse
|
19
|
Paris A, Tardif N, Baietti FM, Berra C, Leclair HM, Leucci E, Galibert M, Corre S. The AhR-SRC axis as a therapeutic vulnerability in BRAFi-resistant melanoma. EMBO Mol Med 2022; 14:e15677. [PMID: 36305167 PMCID: PMC9728058 DOI: 10.15252/emmm.202215677] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 09/30/2022] [Accepted: 09/30/2022] [Indexed: 12/14/2022] Open
Abstract
The nongenetic mechanisms required to control tumor phenotypic plasticity and shape drug-resistance remain unclear. We show here that the Aryl hydrocarbon Receptor (AhR) transcription factor directly regulates the gene expression program associated with the acquisition of resistance to BRAF inhibitor (BRAFi) in melanoma. In addition, we show in melanoma cells that canonical activation of AhR mediates the activation of the SRC pathway and promotes the acquisition of an invasive and aggressive resistant phenotype to front-line BRAFi treatment in melanoma. This nongenetic reprogramming identifies a clinically compatible approach to reverse BRAFi resistance in melanoma. Using a preclinical BRAFi-resistant PDX melanoma model, we demonstrate that SRC inhibition with dasatinib significantly re-sensitizes melanoma cells to BRAFi. Together we identify the AhR/SRC axis as a new therapeutic vulnerability to trigger resistance and warrant the introduction of SRC inhibitors during the course of the treatment in combination with front-line therapeutics to delay BRAFi resistance.
Collapse
Affiliation(s)
- Anaïs Paris
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) – UMR6290, ERL U1305RennesFrance
| | - Nina Tardif
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) – UMR6290, ERL U1305RennesFrance
| | - Francesca M Baietti
- Laboratory for RNA Cancer Biology, Department of OncologyLKI, KU LeuvenLeuvenBelgium,Trace PDX Platform, Department of OncologyLKI, KU LeuvenLeuvenBelgium
| | - Cyrille Berra
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) – UMR6290, ERL U1305RennesFrance,Department of Molecular Genetics and GenomicsHospital University of Rennes (CHU Rennes)RennesFrance
| | - Héloïse M Leclair
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) – UMR6290, ERL U1305RennesFrance
| | - Eleonora Leucci
- Laboratory for RNA Cancer Biology, Department of OncologyLKI, KU LeuvenLeuvenBelgium,Trace PDX Platform, Department of OncologyLKI, KU LeuvenLeuvenBelgium
| | - Marie‐Dominique Galibert
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) – UMR6290, ERL U1305RennesFrance,Department of Molecular Genetics and GenomicsHospital University of Rennes (CHU Rennes)RennesFrance
| | - Sébastien Corre
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) – UMR6290, ERL U1305RennesFrance
| |
Collapse
|
20
|
Romano D, García-Gutiérrez L, Aboud N, Duffy DJ, Flaherty KT, Frederick DT, Kolch W, Matallanas D. Proteasomal down-regulation of the proapoptotic MST2 pathway contributes to BRAF inhibitor resistance in melanoma. Life Sci Alliance 2022; 5:5/10/e202201445. [PMID: 36038253 PMCID: PMC9434705 DOI: 10.26508/lsa.202201445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 11/25/2022] Open
Abstract
The loss of MST2 pathway protein expression in BRAF inhibitor resistant melanoma cells is due to ubiquitination and subsequent proteasomal degradation and prevents MST2-mediated apoptosis. The RAS-RAF-MEK-ERK pathway is hyperactivated in most malignant melanomas, and mutations in BRAF or NRAS account for most of these cases. BRAF inhibitors (BRAFi) are highly efficient for treating patients with BRAFV600E mutations, but tumours frequently acquire resistance within a few months. Multiple resistance mechanisms have been identified, due to mutations or network adaptations that revive ERK signalling. We have previously shown that RAF proteins inhibit the MST2 proapoptotic pathway in a kinase-independent fashion. Here, we have investigated the role of the MST2 pathway in mediating resistance to BRAFi. We show that the BRAFV600E mutant protein, but not the wild-type BRAF protein, binds to MST2 inhibiting its proapoptotic signalling. Down-regulation of MST2 reduces BRAFi-induced apoptosis. In BRAFi-resistant cell lines, MST2 pathway proteins are down-regulated by ubiquitination and subsequent proteasomal degradation rendering cells refractory to MST2 pathway–induced apoptosis. Restoration of apoptosis can be achieved by increasing MST2 pathway protein expression using proteasome inhibitors. In summary, we show that the MST2 pathway plays a role in the acquisition of BRAFi resistance in melanoma.
Collapse
Affiliation(s)
- David Romano
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | | | - Nourhan Aboud
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - David J Duffy
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland.,Department of Biology/Whitney Laboratory for Marine Bioscience, University of Florida, Gainesville, FL, USA
| | | | | | - Walter Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland .,Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - David Matallanas
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
21
|
Zheng P, Zhou C, Lu L, Liu B, Ding Y. Elesclomol: a copper ionophore targeting mitochondrial metabolism for cancer therapy. J Exp Clin Cancer Res 2022; 41:271. [PMID: 36089608 PMCID: PMC9465867 DOI: 10.1186/s13046-022-02485-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/02/2022] [Indexed: 01/06/2023] Open
Abstract
Elesclomol is an anticancer drug that targets mitochondrial metabolism. In the past, elesclomol was recognized as an inducer of oxidative stress, but now it has also been found to suppress cancer by inducing cuproptosis. Elesclomol’s anticancer activity is determined by the dependence of cancer on mitochondrial metabolism. The mitochondrial metabolism of cancer stem cells, cancer cells resistant to platinum drugs, proteasome inhibitors, molecularly targeted drugs, and cancer cells with inhibited glycolysis was significantly enhanced. Elesclomol exhibited tremendous toxicity to all three kinds of cells. Elesclomol's toxicity to cells is highly dependent on its transport of extracellular copper ions, a process involved in cuproptosis. The discovery of cuproptosis has perfected the specific cancer suppressor mechanism of elesclomol. For some time, elesclomol failed to yield favorable results in oncology clinical trials, but its safety in clinical application was confirmed. Research progress on the relationship between elesclomol, mitochondrial metabolism and cuproptosis provides a possibility to explore the reapplication of elesclomol in the clinic. New clinical trials should selectively target cancer types with high mitochondrial metabolism and attempt to combine elesclomol with platinum, proteasome inhibitors, molecularly targeted drugs, or glycolysis inhibitors. Herein, the particular anticancer mechanism of elesclomol and its relationship with mitochondrial metabolism and cuproptosis will be presented, which may shed light on the better application of elesclomol in clinical tumor treatment.
Collapse
|
22
|
Yu J, Wu X, Song J, Zhao Y, Li H, Luo M, Liu X. Loss of MHC-I antigen presentation correlated with immune checkpoint blockade tolerance in MAPK inhibitor-resistant melanoma. Front Pharmacol 2022; 13:928226. [PMID: 36091815 PMCID: PMC9459091 DOI: 10.3389/fphar.2022.928226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/27/2022] [Indexed: 11/20/2022] Open
Abstract
Immune checkpoint blockade and MAPK-targeted combined therapy is a promising regimen for advanced melanoma patients. However, the clinical benefit from this combo regimen remains limited, especially in patients who acquired resistance to MAPK-targeted therapy. Here, we systematically characterized the immune landscape during MAPK-targeted therapy in patients and mouse melanoma models. We observed that both the abundance of tumor-infiltrated T cells and the expression of immune-related genes were upregulated in the drug-responsive period, but downregulated in the resistance period, implying that acquired drug resistance dampens the antitumor immune response. Further transcriptomic dissection indicated that loss of MHC-I antigen presentation on tumor cells plays a critical role in the reduction of T cell infiltration during drug resistance. Survival analysis demonstrates that loss of antigen presentation and reduction of T-cell infiltration during acquired drug resistance are associated with poorer clinical response and prognosis of anti-PD-1 therapy in melanoma patients. In addition, we identified that alterations in the MAPK inhibitor resistance-related oncogenic signaling pathway closely correlated with deficiency of MHC-I antigen presentation, including activation of the PI3K-mTOR, MAPK, and Wnt pathways. In conclusion, our research illuminates that decreased infiltration of T cells is associated with acquired drug resistance during MAPK-targeted therapy, which may underlie the cross-resistance to immune checkpoint blockade.
Collapse
Affiliation(s)
- Jing Yu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Xi Wu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Jinen Song
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Yujie Zhao
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Huifang Li
- Research Core Facility, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Min Luo
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
- *Correspondence: Xiaowei Liu, ; Min Luo,
| | - Xiaowei Liu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
- *Correspondence: Xiaowei Liu, ; Min Luo,
| |
Collapse
|
23
|
Pietrobono S, De Paolo R, Mangiameli D, Marranci A, Battisti I, Franchin C, Arrigoni G, Melisi D, Poliseno L, Stecca B. p38 MAPK-dependent phosphorylation of transcription factor SOX2 promotes an adaptive response to BRAF inhibitors in melanoma cells. J Biol Chem 2022; 298:102353. [PMID: 35944584 PMCID: PMC9463537 DOI: 10.1016/j.jbc.2022.102353] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 10/26/2022] Open
|
24
|
Targeting EGFR in melanoma - The sea of possibilities to overcome drug resistance. Biochim Biophys Acta Rev Cancer 2022; 1877:188754. [PMID: 35772580 DOI: 10.1016/j.bbcan.2022.188754] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/15/2022] [Accepted: 06/23/2022] [Indexed: 12/21/2022]
Abstract
Melanoma is considered one of the most aggressive skin cancers. It spreads and metastasizes quickly and is intrinsically resistant to most conventional chemotherapeutics, thereby presenting a challenge to researchers and clinicians searching for effective therapeutic strategies to treat patients with melanoma. The use of inhibitors of mutated serine/threonine-protein kinase B-RAF (BRAF), e.g., vemurafenib and dabrafenib, has revolutionized melanoma chemotherapy. Unfortunately, the response to these drugs lasts a limited time due to the development of acquired resistance. One of the proteins responsible for this process is epidermal growth factor receptor (EGFR). In this review, we summarize the role of EGFR signaling in the multidrug resistance of melanomas and discuss possible applications of EGFR inhibitors to overcome the development of drug resistance in melanoma cells during therapy.
Collapse
|
25
|
Xu T, Wang X, Wang Z, Deng T, Qi C, Liu D, Li Y, Ji C, Li J, Shen L. Molecular mechanisms underlying the resistance of BRAF V600E-mutant metastatic colorectal cancer to EGFR/BRAF inhibitors. Ther Adv Med Oncol 2022; 14:17588359221105022. [PMID: 35747165 PMCID: PMC9210093 DOI: 10.1177/17588359221105022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 05/16/2022] [Indexed: 11/16/2022] Open
Abstract
Background Combinatorial inhibition of epidermal growth factor receptor (EGFR) and BRAF shows remarkable clinical benefits in patients with BRAF V600E-mutant metastatic colorectal cancer (mCRC). However, the tumor may inevitably develop resistance to the targeted therapy, thereby limiting the response rate and durability. This study aimed to determine the genetic alterations associated with intrinsic and acquired resistance to EGFR/BRAF inhibitors in BRAF V600E-mutant mCRC. Methods Targeted sequencing of 520 cancer-related genes was performed in tumor tissues and in plasma samples collected from patients with BRAF V600E-mutant mCRC, who were treated with EGFR/BRAF ± MEK inhibitors, before and after the targeted treatment. Clinical benefit was defined as an objective response or a stable disease lasting longer than the median progression-free survival (PFS). Results In all, 25 patients with BRAF V600E-mutant mCRC were included in this study. Those with RNF43 mutations (n = 8) were more likely to achieve clinical benefit from EGFR/BRAF inhibitors than those with wild-type RNF43 (87.5% versus 37.5%, p = 0.034). Genetic alterations in receptor tyrosine kinase genes (n = 6) were associated with worse PFS (p = 0.005). Among the 23 patients whose disease progressed after the EGFR/BRAF-targeted therapy, at least one acquired resistance-related mutation was detected in 12 patients. Acquired mutations were most frequently observed in the mitogen-activated protein kinase pathway-related genes (n = 9), including KRAS (G12D and Q61H/R), NRAS (Q61L/R/K and amplification), BRAF (amplification), and MEK1 (K57T). MET amplification and PIK3R1 Q579fs mutation emerged in three patients and one patient, respectively, after disease progression. Conclusion Multiple genetic alterations are associated with clinical benefits and resistance to EGFR/BRAF inhibitors in BRAF V600E-mutant mCRC. Our findings provide novel insights into strategies for overcoming resistance to EGFR/BRAF inhibitors in patients with BRAF V600E-mutant mCRC.
Collapse
Affiliation(s)
- Ting Xu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xicheng Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Ting Deng
- National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Changsong Qi
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Dan Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanyan Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Congcong Ji
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| |
Collapse
|
26
|
Dzung A, Saltari A, Tiso N, Lyck R, Dummer R, Levesque MP. STK11 Prevents Invasion through Signal Transducer and Activator of Transcription 3/5 and FAK Repression in Cutaneous Melanoma. J Invest Dermatol 2022; 142:1171-1182.e10. [PMID: 34757069 DOI: 10.1016/j.jid.2021.09.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 11/18/2022]
Abstract
The STK11/LKB1 is a tumor suppressor involved in metabolism and cell motility. In BRAFV600E melanoma, STK11 is inactivated by extracellular signal‒regulated kinase and RSK, preventing it from binding and activating adenosine monophosphate-activated protein kinase and promoting melanoma cell proliferation. Although STK11 mutations occur in 5‒10% of cutaneous melanoma, few functional studies have been performed. By knocking out STK11 with CRISPR/Cas9 in two human BRAF-mutant melanoma cell lines, we found that STK11 loss reduced the sensitivity to a BRAF inhibitor. More strikingly, STK11 loss led to an increased invasive phenotype in both three-dimensional spheroids and in vivo zebrafish xenograft models. STK11 overexpression consistently reversed the invasive phenotype. Interestingly, STK11 knockout increased invasion also in an NRAS-mutant melanoma cell line. Furthermore, although STK11 was expressed in primary human melanoma tumors, its expression significantly decreased in melanoma metastases, especially in brain metastases. In the STK11-knockout cells, we observed increased activating phosphorylation of signal transducer and activator of transcription 3/5 and FAK. Using inhibitors of signal transducer and activator of transcription 3/5 and FAK, we reversed the invasive phenotype in both BRAF- and NRAS-mutated cells. Our findings confirm an increased invasive phenotype on STK11 inactivation in BRAF- and NRAS-mutant cutaneous melanoma that can be targeted by signal transducer and activator of transcription 3/5 and FAK inhibition.
Collapse
Affiliation(s)
- Andreas Dzung
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Annalisa Saltari
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Natascia Tiso
- Laboratory of Developmental Genetics, Department of Biology, University of Padova, Padova, Italy
| | - Ruth Lyck
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
27
|
Adams R, Coumbe JEM, Coumbe BGT, Thomas J, Willsmore Z, Dimitrievska M, Yasuzawa-Parker M, Hoyle M, Ingar S, Geh J, MacKenzie Ross A, Healy C, Papa S, Lacy KE, Karagiannis SN. BRAF inhibitors and their immunological effects in malignant melanoma. Expert Rev Clin Immunol 2022; 18:347-362. [PMID: 35195495 DOI: 10.1080/1744666x.2022.2044796] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The treatment of cutaneous melanoma has been revolutionised by the development of small molecule inhibitors targeting the MAPK pathway, including inhibitors of BRAF (BRAFi) and MEK (MEKi), and immune checkpoint blockade antibodies, occurring in tandem. Despite these advances, the 5-year survival rate for patients with advanced melanoma remains only around 50%. Although not designed to alter immune responses within the tumour microenvironment (TME), MAPK pathway inhibitors (MAPKi) exert a range of effects on the host immune compartment which may offer opportunities for therapeutic interventions. AREAS COVERED We review the effects of MAPKi especially BRAFi, on the TME, focussing on alterations in inflammatory cytokine secretion, the recruitment of immune cells and their functions, both during response to BRAFi treatment and as resistance develops. We outline potential combinations of MAPKi with established and experimental treatments. EXPERT OPINION MAPKi in combination or in sequence with established treatments such as checkpoint inhibitors, anti-angiogenic agents, or new therapies such as adoptive cell therapies, may augment their immunological effects, reverse tumour-associated immune suppression and offer the prospect of longer-lived clinical responses. Refining therapeutic tools at our disposal and embracing "old friends" in the melanoma treatment arsenal, alongside new target identification, may improve the chances of therapeutic success.
Collapse
Affiliation(s)
- Rebecca Adams
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Jack E M Coumbe
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Ben G T Coumbe
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Jennifer Thomas
- The Royal Marsden, Downs Road, Sutton, Surrey, United Kingdom
| | - Zena Willsmore
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Marija Dimitrievska
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Monica Yasuzawa-Parker
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Maximilian Hoyle
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Suhaylah Ingar
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Jenny Geh
- Department of Plastic Surgery at Guy's, King's, and St. Thomas' Hospitals, London, United Kingdom
| | - Alastair MacKenzie Ross
- Department of Plastic Surgery at Guy's, King's, and St. Thomas' Hospitals, London, United Kingdom
| | - Ciaran Healy
- Department of Plastic Surgery at Guy's, King's, and St. Thomas' Hospitals, London, United Kingdom
| | - Sophie Papa
- Department of Medical Oncology, Guy's and St. Thomas' NHS Foundation Trust, London, United Kingdom.,ImmunoEngineering, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Katie E Lacy
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom.,Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London SE1 9RT, United Kingdom
| |
Collapse
|
28
|
de Groot E, Varghese S, Tan L, Knighton B, Sobieski M, Nguyen N, Park YS, Powell R, Lorenzi PL, Zheng B, Stephan C, Gopal YNV. Combined inhibition of HMGCoA reductase and mitochondrial complex I induces tumor regression of BRAF inhibitor-resistant melanomas. Cancer Metab 2022; 10:6. [PMID: 35193687 PMCID: PMC8862475 DOI: 10.1186/s40170-022-00281-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/10/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Primary and posttreatment resistance to BRAFV600 mutation-targeting inhibitors leads to disease relapse in a majority of melanoma patients. In many instances, this resistance is promoted by upregulation of mitochondrial oxidative phosphorylation (OxPhos) in melanoma cells. We recently showed that a novel electron transport chain (ETC) complex I inhibitor, IACS-010759 (IACS), abolished OxPhos and significantly inhibited tumor growth of high-OxPhos, BRAF inhibitor (BRAFi)-resistant human melanomas. However, the inhibition was not uniform across different high OxPhos melanomas, and combination with BRAFi did not improve efficacy. METHODS We performed a high-throughput unbiased combinatorial drug screen of clinically relevant small molecules to identify the most potent combination agent with IACS for inhibiting the growth of high-OxPhos, BRAFi-resistant melanomas. We performed bioenergetics and carbon-13 metabolite tracing to delineate the metabolic basis of sensitization of melanomas to the combination treatment. We performed xenograft tumor growth studies and Reverse-Phase Protein Array (RPPA)-based functional proteomics analysis of tumors from mice fed with regular or high-fat diet to evaluate in vivo molecular basis of sensitization to the combination treatment. RESULTS A combinatorial drug screen and subsequent validation studies identified Atorvastatin (STN), a hydroxymethylglutaryl-coenzyme A reductase inhibitor (HMGCRi), as the most potent treatment combination with IACS to inhibit in vitro cell growth and induce tumor regression or stasis of some BRAFi-resistant melanomas. Bioenergetics analysis revealed a dependence on fatty acid metabolism in melanomas that responded to the combination treatment. RPPA analysis and carbon-13 tracing analysis in these melanoma cells showed that IACS treatment decreased metabolic fuel utilization for fatty acid metabolism, but increased substrate availability for activation of the mevalonate pathway by HMGCR, creating a dependence on this pathway. Functional proteomic analysis showed that IACS treatment inhibited MAPK but activated AKT pathway. Combination treatment with STN counteracted AKT activation. CONCLUSIONS STN and other clinically approved HMGCRi could be promising combinatorial agents for improving the efficacy of ETC inhibitors like IACS in BRAFi-resistant melanomas.
Collapse
Affiliation(s)
- Evelyn de Groot
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Sruthy Varghese
- Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Lin Tan
- Department of Bioinformatics and Computational Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Barbara Knighton
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Mary Sobieski
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Nghi Nguyen
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Yong Sung Park
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Reid Powell
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Bin Zheng
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Clifford Stephan
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Y N Vashisht Gopal
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
- Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
29
|
Hu L, Fan M, Shi S, Song X, Wang F, He H, Qi B. Dual target inhibitors based on EGFR: Promising anticancer agents for the treatment of cancers (2017-). Eur J Med Chem 2022; 227:113963. [PMID: 34749202 DOI: 10.1016/j.ejmech.2021.113963] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/23/2021] [Accepted: 10/28/2021] [Indexed: 02/06/2023]
Abstract
The EGFR family play a significant role in cell signal transduction and their overexpression is implicated in the pathogenesis of numerous human solid cancers. Inhibition of the EGFR-mediated signaling pathways by EGFR inhibitors is a widely used strategy for the treatment of cancers. In most cases, the EGFR inhibitors used in clinic were only effective when the cancer cells harbored specific activating EGFR mutations which appeared to preserve the ligand-dependency of receptor activation but altered the pattern of downstream signaling pathways. Moreover, cancer is a kind of multifactorial disease, and therefore manipulating a single target may result in treatment failure. Although drug combinations for the treatment of cancers proved to be successful, the use of two or more drugs concurrently still was a challenge in clinical therapy owing to various dose-limiting toxicities and drug-drug interactions caused by pharmacokinetic profiles changed. Therefore, a single drug targeting two or multiple targets could serve as an effective strategy for the treatment of cancers. In recent, drugs with diverse pharmacological effects have been shown to be more advantageous than combination therapies due to their lower incidences of side effects and more resilient therapies. Accordingly, dual target-single-agent strategy has become a popular field for cancer treatment, and researchers became more and more interest in the development of novel dual-target drugs in recent years. In this review, we briefly introduce the EGFR family proteins and synergisms between EGFR and other anticancer targets, and summarizes the development of potential dual target inhibitors based on wild-type and/or mutant EGFR for the treatment of solid cancers in the past five years. Additionally, the rational design and SARs of these dual target agents are also presented in detailed, which will lay a significant foundation for the further development of novel EGFR-based dual inhibitors with excellent druggability.
Collapse
Affiliation(s)
- Liping Hu
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Mengmeng Fan
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Shengmin Shi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Xiaomeng Song
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Fei Wang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Huan He
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China.
| | - Baohui Qi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China.
| |
Collapse
|
30
|
Kohtamäki L, Arjama M, Mäkelä S, Ianevski P, Välimäki K, Juteau S, Ilmonen S, Ungureanu D, Kallioniemi O, Murumägi A, Hernberg M. High-throughput ex vivo drug testing identifies potential drugs and drug combinations for NRAS-positive malignant melanoma. Transl Oncol 2022; 15:101290. [PMID: 34837846 PMCID: PMC8633005 DOI: 10.1016/j.tranon.2021.101290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/05/2021] [Accepted: 11/17/2021] [Indexed: 12/18/2022] Open
Abstract
Therapy options for patients with metastatic melanoma (MM) have considerably improved over the past decade. However, many patients still need effective therapy after unsuccessful immunotherapy, especially patients with BRAF-negative tumors who lack the option of targeted treatment second line. Therefore, the elucidation of efficient and personalized therapy options for these patients is required. In this study, three patient-derived cancer cells (PDCs) were established from NRAS Q61-positive MM patients. The response of PDCs and five established melanoma cell lines (two NRAS-positive, one wild type, and two BRAF V600-positive) was evaluated toward a panel of 527 oncology drugs using high-throughput drug sensitivity and resistance testing. The PDCs and cell lines displayed strong responses to MAPK inhibitors, as expected. Additionally, the PDCs and cell lines were responsive to PI3K/mTOR, mTOR, and PLK1 inhibitors among other effective drugs currently undergoing clinical trials. Combinations with a MEK inhibitor were tested with other targeted agents to identify effective synergies. MEK inhibitor showed synergy with multikinase inhibitor ponatinib, ABL inhibitor nilotinib, PI3K/mTOR inhibitor pictilisib, and pan-RAF inhibitor LY3009120. The application of the patients' cancer cells for functional drug testing ex vivo is one step further in the process of identifying potential agents and agent combinations to personalize treatment for patients with MM. Our preliminary study results suggest that this approach has the potential for larger-scale drug testing and personalized treatment applications in our expansion trial. Our results show that drug sensitivity and resistance testing may be implementable in the treatment planning of patients with MM.
Collapse
Affiliation(s)
- Laura Kohtamäki
- Helsinki University Hospital, Comprehensive Cancer Center, Department of Oncology, Helsinki and University of Helsinki, Finland.
| | - Mariliina Arjama
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), Helsinki, Finland and University of Helsinki, Finland
| | - Siru Mäkelä
- Helsinki University Hospital, Comprehensive Cancer Center, Department of Oncology, Helsinki and University of Helsinki, Finland
| | - Philipp Ianevski
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), Helsinki, Finland and University of Helsinki, Finland
| | - Katja Välimäki
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), Helsinki, Finland and University of Helsinki, Finland
| | - Susanna Juteau
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Suvi Ilmonen
- Helsinki University Hospital, Department of Surgery, Helsinki and University of Helsinki, Finland
| | - Daniela Ungureanu
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), Helsinki, Finland and University of Helsinki, Finland; Science for Life Laboratory (SciLifeLab), Department of Oncology and Pathology, Karolinska Institutet, Sweden
| | - Astrid Murumägi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), Helsinki, Finland and University of Helsinki, Finland.
| | - Micaela Hernberg
- Helsinki University Hospital, Comprehensive Cancer Center, Department of Oncology, Helsinki and University of Helsinki, Finland
| |
Collapse
|
31
|
Dent P, Booth L, Poklepovic A, Kirkwood JM. Neratinib kills B-RAF V600E melanoma via ROS-dependent autophagosome formation and death receptor signaling. Pigment Cell Melanoma Res 2022; 35:66-77. [PMID: 34482636 PMCID: PMC11559083 DOI: 10.1111/pcmr.13014] [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: 05/22/2021] [Revised: 08/02/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022]
Abstract
Melanoma cells expressing mutant B-RAF V600E are susceptible to treatment with the combination of a B-RAF inhibitor and a MEK1/2 inhibitor. We investigated the impact of the ERBB family and MAP4K inhibitor neratinib on the biology of PDX isolates of cutaneous melanoma expressing B-RAF V600E. Neratinib synergized with HDAC inhibitors to kill melanoma cells at their physiologic concentrations. Neratinib activated ATM, AMPK, ULK1, and PERK and inactivated mTORC1/2, ERK1/2, eIF2 alpha, and STAT3. Neratinib increased expression of Beclin1, ATG5, CD95, and FAS-L and decreased levels of multiple toxic BH3 domain proteins, MCL1, BCL-XL, FLIP-s, and ERBB1/2/4. ATG13 S318 phosphorylation and autophagosome formation was dependent upon ATM, and activation of ATM was dependent on reactive oxygen species. Reduced expression of ERBB1/2/4 required autophagosome formation and reduced MCL1/BCL-XL levels required eIF2 alpha phosphorylation. Maximal levels of eIF2 alpha phosphorylation required signaling by ATM-AMPK and autophagosome formation. Knock down of eIF2 alpha, CD95, FAS-L, Beclin1, and ATG5 or over-expression of FLIP-s significantly reduced killing. Combined knock down of Beclin1 and CD95 abolished cell death. Our data demonstrate that PDX melanoma cells expressing B-RAF V600E are susceptible to being killed by neratinib and more so when combined with HDACi.
Collapse
Affiliation(s)
- Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Andrew Poklepovic
- Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - John M. Kirkwood
- Melanoma and Skin Cancer Program, Hillman Cancer Research Pavilion Laboratory, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
32
|
Olmeda D, Cerezo‐Wallis D, Mucientes C, Calvo TG, Cañón E, Alonso‐Curbelo D, Ibarz N, Muñoz J, Rodriguez‐Peralto JL, Ortiz‐Romero P, Ortega S, Soengas MS. Live imaging of neolymphangiogenesis identifies acute antimetastatic roles of dsRNA mimics. EMBO Mol Med 2021; 13:e12924. [PMID: 34762341 PMCID: PMC8649872 DOI: 10.15252/emmm.202012924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 11/18/2022] Open
Abstract
Long-range communication between tumor cells and the lymphatic vasculature defines competency for metastasis in different cancer types, particularly in melanoma. Nevertheless, the discovery of selective blockers of lymphovascular niches has been compromised by the paucity of experimental systems for whole-body analyses of tumor progression. Here, we exploit immunocompetent and immunodeficient mouse models for live imaging of Vegfr3-driven neolymphangiogenesis, as a versatile platform for drug screening in vivo. Spatiotemporal analyses of autochthonous melanomas and patient-derived xenografts identified double-stranded RNA mimics (dsRNA nanoplexes) as potent inhibitors of neolymphangiogenesis, metastasis, and post-surgical disease relapse. Mechanistically, dsRNA nanoplexes were found to exert a rapid dual action in tumor cells and in their associated lymphatic vasculature, involving the transcriptional repression of the lymphatic drivers Midkine and Vegfr3, respectively. This suppressive function was mediated by a cell-autonomous type I interferon signaling and was not shared by FDA-approved antimelanoma treatments. These results reveal an alternative strategy for targeting the tumor cell-lymphatic crosstalk and underscore the power of Vegfr3-lymphoreporters for pharmacological testing in otherwise aggressive cancers.
Collapse
Affiliation(s)
- David Olmeda
- Melanoma LaboratoryMolecular Oncology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Daniela Cerezo‐Wallis
- Melanoma LaboratoryMolecular Oncology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
- Present address:
Spanish National Center for Cardiovascular Research (CNIC)MadridSpain
| | - Cynthia Mucientes
- Melanoma LaboratoryMolecular Oncology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Tonantzin G Calvo
- Melanoma LaboratoryMolecular Oncology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Estela Cañón
- Melanoma LaboratoryMolecular Oncology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Direna Alonso‐Curbelo
- Melanoma LaboratoryMolecular Oncology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
- Present address:
Memorial Sloan Kettering Cancer CentreNew YorkNYUSA
| | - Nuria Ibarz
- Proteomics UnitBiotechnology Programme, ProteoRed‐ISCIIISpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Javier Muñoz
- Proteomics UnitBiotechnology Programme, ProteoRed‐ISCIIISpanish National Cancer Research Centre (CNIO)MadridSpain
| | - José L Rodriguez‐Peralto
- Instituto de Investigación i+12Hospital 12 de OctubreUniversidad Complutense Madrid Medical SchoolMadridSpain
| | - Pablo Ortiz‐Romero
- Department of DermatologyHospital 12 de OctubreUniversidad Complutense Madrid Medical SchoolMadridSpain
| | - Sagrario Ortega
- Mouse Genome Editing Core UnitSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - María S Soengas
- Melanoma LaboratoryMolecular Oncology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
| |
Collapse
|
33
|
Zhang X, Wang W, Wang Y, Jiang G. Identification of genes and pathways leading to metastasis and poor prognosis in melanoma. Aging (Albany NY) 2021; 13:22474-22489. [PMID: 34582363 PMCID: PMC8507267 DOI: 10.18632/aging.203554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 09/03/2021] [Indexed: 01/08/2023]
Abstract
Melanoma causes the highest mortality rate among all skin cancers. However, the underlying molecular mechanisms leading to metastasis and poor prognosis in melanoma have not been fully elucidated. In this study, the differentially expressed genes (DEGs) related to metastasis in melanoma were screened out. The results of gene annotation was combined with The Cancer Genome Atlas (TCGA) database. The microRNA (miRNA) network that regulates key genes and their correlation with BRAFV600E was preliminarily analyzed. Cell and molecular biology experiments were conducted to verify the results of bioinformatics analysis. Results showed that the PI3K-Akt signaling pathway contained the key genes CDK2, CDK4, KIT, and Von Willebrand factor. Survival analysis showed that high expression of the four key genes significantly reduced the survival rate of patients with melanoma. Correlation analysis showed that BRAFV600E may regulate the expression of the four key genes, and a total of 240 miRNAs may regulate this expression. Experiments showed that the inactivation of key genes inhibits the proliferation, migration, and invasion of melanoma. In conclusion, the PI3K-Akt signaling pathway and the four key genes promoted the proliferation, migration, and invasion of melanoma, and related to poor prognosis of patients with melanoma.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Dermatology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Xuzhou Medical University, Xuzhou, China
| | - Wandong Wang
- Department of Dermatology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Xuzhou Medical University, Xuzhou, China
| | - Yun Wang
- Department of Dermatology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Xuzhou Medical University, Xuzhou, China
| | - Guan Jiang
- Department of Dermatology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
34
|
A Novel Regimen for Treating Melanoma: MCL1 Inhibitors and Azacitidine. Pharmaceuticals (Basel) 2021; 14:ph14080749. [PMID: 34451846 PMCID: PMC8399604 DOI: 10.3390/ph14080749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/19/2021] [Accepted: 07/23/2021] [Indexed: 12/22/2022] Open
Abstract
Although treatment options for melanoma patients have expanded in recent years with the approval of immunotherapy and targeted therapy, there is still an unmet need for new treatment options for patients that are ineligible for, or resistant to these therapies. BH3 mimetics, drugs that mimic the activity of pro-apoptotic BCL2 family proteins, have recently achieved remarkable success in the clinical setting. The combination of BH3 mimetic ABT-199 (venetoclax) plus azacitidine has shown substantial benefit in treating acute myelogenous leukemia. We evaluated the efficacy of various combinations of BH3 mimetic + azacitidine in fourteen human melanoma cell lines from cutaneous, mucosal, acral and uveal subtypes. Using a combination of cell viability assay, BCL2 family knockdown cell lines, live cell imaging, and sphere formation assay, we found that combining inhibition of MCL1, an anti-apoptotic BCL2 protein, with azacitidine had substantial pro-apoptotic effects in multiple melanoma cell lines. Specifically, this combination reduced cell viability, proliferation, sphere formation, and induced apoptosis. In addition, this combination is highly effective at reducing cell viability in rare mucosal and uveal subtypes. Overall, our data suggest this combination as a promising therapeutic option for some patients with melanoma and should be further explored in clinical trials.
Collapse
|
35
|
Stagno A, Vari S, Annovazzi A, Anelli V, Russillo M, Cognetti F, Ferraresi V. Case Report: Rechallenge With BRAF and MEK Inhibitors in Metastatic Melanoma: A Further Therapeutic Option in Salvage Setting? Front Oncol 2021; 11:645008. [PMID: 34136385 PMCID: PMC8202400 DOI: 10.3389/fonc.2021.645008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/11/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The combination of BRAF and MEK inhibitors represents the standard of care treatment for patients with metastatic BRAF-mutated melanoma, notwithstanding the high frequency of emergent resistance. Moreover, therapeutic options outside clinical trials are scarce when patients have progressed after both targeted therapy and therapy with immune checkpoint inhibitors. In this article, we report our experience with targeted therapy rechallenging with BRAF and MEK inhibitors in patients with metastatic BRAF-mutated melanoma after progression with kinase inhibitors and immunotherapy. METHODS Four patients with metastatic BRAF-mutated melanoma were rechallenged with BRAF and MEK inhibitors after progression with targeted therapy and subsequent immunotherapy (checkpoint inhibitors). RESULTS Two patients (one of them was heavily pretreated) had partial response over 36 months (with local treatment on oligoprogression disease) and 10 months, respectively. A third patient with multisite visceral disease and high serum levels of lactate dehydrogenase had a short-lived clinical benefit rapidly followed by massive progression of disease (early progressor). The fourth patient, currently on treatment with BRAF/MEK inhibitors, is showing a clinical benefit and radiological stable disease over 3 months of therapy. Adverse events were manageable, similar to those reported during the first targeted therapy; the treatment was better tolerated at rechallenge compared with the first treatment by two out of four patients.
Collapse
Affiliation(s)
- Anna Stagno
- Department of Medical Oncology 1, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Sabrina Vari
- Department of Medical Oncology 1, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Alessio Annovazzi
- Nuclear Medicine Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Vincenzo Anelli
- Radiology and Diagnostic Imaging Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Michelangelo Russillo
- Department of Medical Oncology 1, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Francesco Cognetti
- Department of Medical Oncology 1, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Virginia Ferraresi
- Department of Medical Oncology 1, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
36
|
de Souza N, de Oliveira ÉA, Faião-Flores F, Pimenta LA, Quincoces JAP, Sampaio SC, Maria-Engler SS. Metalloproteinases Suppression Driven by the Curcumin Analog DM-1 Modulates Invasion in BRAF-Resistant Melanomas. Anticancer Agents Med Chem 2021; 20:1038-1050. [PMID: 32067622 DOI: 10.2174/1871520620666200218111422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/15/2020] [Accepted: 01/31/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Melanoma is the most aggressive skin cancer, and BRAF (V600E) is the most frequent mutation that led to the development of BRAF inhibitors (BRAFi). However, patients treated with BRAFi usually present recidivism after 6-9 months. Curcumin is a turmeric substance, and it has been deeply investigated due to its anti-inflammatory and antitumoral effects. Still, the low bioavailability and biodisponibility encouraged the investigation of different analogs. DM-1 is a curcumin analog and has shown an antitumoral impact in previous studies. METHODS Evaluated DM-1 stability and cytotoxic effects for BRAFi-sensitive and resistant melanomas, as well as the role in the metalloproteinases modulation. RESULTS DM-1 showed growth inhibitory potential for melanoma cells, demonstrated by reduction of colony formation, migration and endothelial tube formation, and cell cycle arrest. Subtoxic doses were able to downregulate important Metalloproteinases (MMPs) related to invasiveness, such as MMP-1, -2 and -9. Negative modulations of TIMP-2 and MMP-14 reduced MMP-2 and -9 activity; however, the reverse effect is seen when increased TIMP-2 and MMP-14 resulted in raised MMP-2. CONCLUSION These findings provide essential details into the functional role of DM-1 in melanomas, encouraging further studies in the development of combinatorial treatments for melanomas.
Collapse
Affiliation(s)
- Nayane de Souza
- Skin Biology Group, Clinical Chemistry and Toxicology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Brazil
| | - Érica Aparecida de Oliveira
- Skin Biology Group, Clinical Chemistry and Toxicology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Brazil
| | - Fernanda Faião-Flores
- Skin Biology Group, Clinical Chemistry and Toxicology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Brazil
| | | | - José A P Quincoces
- Laboratory of Organic Synthesis, Anhanguera University of São Paulo, UNIAN, Sao Paulo, Brazil
| | - Sandra C Sampaio
- Butantan Institute, Pathophysiology Laboratory, Sao Paulo, Brazil
| | - Silvya S Maria-Engler
- Skin Biology Group, Clinical Chemistry and Toxicology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Brazil
| |
Collapse
|
37
|
Fukushima H, Iwata Y, Saito K, Sugiura K. Successful rechallenge therapy for BRAF/MEK inhibitor-resistant multiple brain metastases of melanoma. J Dermatol 2021; 48:1291-1295. [PMID: 34018641 DOI: 10.1111/1346-8138.15969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/06/2021] [Accepted: 04/20/2021] [Indexed: 11/30/2022]
Abstract
Combination therapy with BRAF and MEK inhibitors (BRAFi/MEKi) have dramatically improved prognosis among patients with BRAF-mutant metastatic melanoma compared with traditional treatment, such as chemotherapy. However, resistance to these targeted agents occurs invariably, thereby limiting their clinical efficacy. Recently, it has been reported that the ligand-independent phosphorylation of erythropoietin-producing hepatocellular receptor A2 (EphA2) at Ser-897 signaling is a driver of BRAF inhibitor resistance in melanoma. A melanoma patient with multiple metastases was treated with dabrafenib plus trametinib therapy and maintained complete remission for more than 2 years. As brain metastasis occurred, we had switched to nivolumab plus ipilimumab therapy. However, new lesions were observed after four cycles of nivolumab plus ipilimumab therapy, she was rechallenged with encorafenib plus binimetinib therapy, and she maintained progression-free status for more than 7 months. We performed immunohistochemical staining of EphA2, phospho-EphA2 (p-EphA2; Ser-897), and epidermal growth factor receptor (EGFR) of melanoma cells before and/or after dabrafenib and trametinib therapy. Immunohistochemical examination showed higher expression of EphA2, p-EphA2, and EGFR in the melanoma cells after dabrafenib plus trametinib therapy as compared with that before therapy. Our results may indicate that EphA2, p-EphA2, and EGFR can be critical factors for resistance and reversible response of BRAFi/MEKi in metastases of melanoma. Our case presents a possible treatment that can help overcome BRAFi/MEKi resistance and improve prognosis of melanoma.
Collapse
Affiliation(s)
- Hidehiko Fukushima
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yohei Iwata
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Kenta Saito
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Kazumitsu Sugiura
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| |
Collapse
|
38
|
McKenna S, García-Gutiérrez L. Resistance to Targeted Therapy and RASSF1A Loss in Melanoma: What Are We Missing? Int J Mol Sci 2021; 22:5115. [PMID: 34066022 PMCID: PMC8150731 DOI: 10.3390/ijms22105115] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/26/2021] [Accepted: 05/06/2021] [Indexed: 12/20/2022] Open
Abstract
Melanoma is one of the most aggressive forms of skin cancer and is therapeutically challenging, considering its high mutation rate. Following the development of therapies to target BRAF, the most frequently found mutation in melanoma, promising therapeutic responses were observed. While mono- and combination therapies to target the MAPK cascade did induce a therapeutic response in BRAF-mutated melanomas, the development of resistance to MAPK-targeted therapies remains a challenge for a high proportion of patients. Resistance mechanisms are varied and can be categorised as intrinsic, acquired, and adaptive. RASSF1A is a tumour suppressor that plays an integral role in the maintenance of cellular homeostasis as a central signalling hub. RASSF1A tumour suppressor activity is commonly lost in melanoma, mainly by aberrant promoter hypermethylation. RASSF1A loss could be associated with several mechanisms of resistance to MAPK inhibition considering that most of the signalling pathways that RASSF1A controls are found to be altered targeted therapy resistant melanomas. Herein, we discuss resistance mechanisms in detail and the potential role for RASSF1A reactivation to re-sensitise BRAF mutant melanomas to therapy.
Collapse
Affiliation(s)
| | - Lucía García-Gutiérrez
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland;
| |
Collapse
|
39
|
Many Distinct Ways Lead to Drug Resistance in BRAF- and NRAS-Mutated Melanomas. Life (Basel) 2021; 11:life11050424. [PMID: 34063141 PMCID: PMC8148104 DOI: 10.3390/life11050424] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/23/2021] [Accepted: 04/30/2021] [Indexed: 11/17/2022] Open
Abstract
Advanced melanoma is a relentless tumor with a high metastatic potential. The combat of melanoma by using the targeted therapy is impeded because several major driver mutations fuel its growth (predominantly BRAF and NRAS). Both these mutated oncogenes strongly activate the MAPK (MEK/ERK) pathway. Therefore, specific inhibitors of these oncoproteins or MAPK pathway components or their combination have been used for tumor eradication. After a good initial response, resistant cells develop almost universally and need the drug for further expansion. Multiple mechanisms, sometimes very distant from the MAPK pathway, are responsible for the development of resistance. Here, we review many of the mechanisms causing resistance and leading to the dismal final outcome of mutated BRAF and NRAS therapy. Very heterogeneous events lead to drug resistance. Due to this, each individual mechanism would be in fact needed to be determined for a personalized therapy to treat patients more efficiently and causally according to molecular findings. This procedure is practically impossible in the clinic. Other approaches are therefore needed, such as combined treatment with more drugs simultaneously from the beginning of the therapy. This could eradicate tumor cells more rapidly and greatly diminish the possibility of emerging mechanisms that allow the evolution of drug resistance.
Collapse
|
40
|
Overcoming the Challenges of High Quality RNA Extraction from Core Needle Biopsy. Biomolecules 2021; 11:biom11050621. [PMID: 33922016 PMCID: PMC8143498 DOI: 10.3390/biom11050621] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
The use of gene expression profiling (GEP) in cancer management is rising, as GEP can be used for disease classification and diagnosis, tailoring treatment to underlying genetic determinants of pharmacological response, monitoring of therapy response, and prognosis. However, the reliability of GEP heavily depends on the input of RNA in sufficient quantity and quality. This highlights the need for standard procedures to ensure best practices for RNA extraction from often small tumor biopsies with variable tissue handling. We optimized an RNA extraction protocol from fresh-frozen (FF) core needle biopsies (CNB) from breast cancer patients and from formalin-fixed paraffin-embedded (FFPE) tissue when FF CNB did not yield sufficient RNA. Methods to avoid ribonucleases andto homogenize or to deparaffinize tissues and the impact of tissue composition on RNA extraction were studied. Additionally, RNA’s compatibility with the nanoString nCounter® technology was studied. This technology platform enables GEP using small RNA fragments. After optimization of the protocol, RNA of high quality and sufficient quantity was obtained from FF CNB in 92% of samples. For the remaining 8% of cases, FFPE material prepared by the pathology department was used for RNA extraction. Both resulting RNA end products are compatible with the nanoString nCounter® technology.
Collapse
|
41
|
Booth L, West C, Von Hoff D, Kirkwood JM, Dent P. GZ17-6.02 Interacts With [MEK1/2 and B-RAF Inhibitors] to Kill Melanoma Cells. Front Oncol 2021; 11:656453. [PMID: 33898322 PMCID: PMC8061416 DOI: 10.3389/fonc.2021.656453] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
We defined the lethal interaction between the novel therapeutic GZ17-6.02 and the standard of care combination of the MEK1/2 inhibitor trametinib and the B-RAF inhibitor dabrafenib in PDX isolates of cutaneous melanoma expressing a mutant B-RAF V600E protein. GZ17-6.02 interacted with trametinib/dabrafenib in an additive fashion to kill melanoma cells. Regardless of prior vemurafenib resistance, the drugs when combined interacted to prolong ATM S1981/AMPK T172 and eIF2α S51 phosphorylation and prolong the reduced phosphorylation of JAK2 Y1007, STAT3 Y705 and STAT5 Y694. In vemurafenib-resistant cells GZ17-6.02 caused a prolonged reduction in mTORC1 S2448, mTORC2 S2481 and ULK1 S757 phosphorylation; regardless of vemurafenib resistance, GZ17-6.02 caused a prolonged elevation in CD95 and FAS-L expression. Knock down of eIF2α, Beclin1, ATG5, ATM, AMPKα, CD95 or FADD significantly reduced the ability of GZ17-6.02 to kill as a single agent or when combined with the kinase inhibitors. Expression of activated mTOR, activated STAT3, activated MEK1 or activated AKT significantly reduced the ability of GZ17-6.02 to kill as a single agent or when combined with kinase inhibitors; protective effects that were significantly less pronounced in cells treated with trametinib/dabrafenib. Regardless of vemurafenib resistance, the drugs alone or in combination all reduced the expression of PD-L1 and increased the levels of MHCA, which was linked to degradation of multiple HDAC proteins. Our findings support the use of GZ17-6.02 in combination with trametinib/dabrafenib in the treatment of melanomas expressing mutant B-RAF V600E proteins.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Cameron West
- Genzada Pharmaceuticals, Sterling, KS, United States
| | - Daniel Von Hoff
- Translational Genomics Research Institute (TGEN), Phoenix, AZ, United States
| | - John M Kirkwood
- Melanoma and Skin Cancer Program, Hillman Cancer Research Pavilion Laboratory, University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
42
|
Han J, Jung Y, Jun Y, Park S, Lee S. Elucidating molecular mechanisms of acquired resistance to BRAF inhibitors in melanoma using a microfluidic device and deep sequencing. Genomics Inform 2021; 19:e2. [PMID: 33840166 PMCID: PMC8042304 DOI: 10.5808/gi.20074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/09/2021] [Accepted: 01/15/2021] [Indexed: 12/01/2022] Open
Abstract
BRAF inhibitors (e.g., vemurafenib) are widely used to treat metastatic melanoma with the BRAF V600E mutation. The initial response is often dramatic, but treatment resistance leads to disease progression in the majority of cases. Although secondary mutations in the mitogen-activated protein kinase signaling pathway are known to be responsible for this phenomenon, the molecular mechanisms governing acquired resistance are not known in more than half of patients. Here we report a genome- and transcriptome-wide study investigating the molecular mechanisms of acquired resistance to BRAF inhibitors. A microfluidic chip with a concentration gradient of vemurafenib was utilized to rapidly obtain therapy-resistant clones from two melanoma cell lines with the BRAF V600E mutation (A375 and SK-MEL-28). Exome and transcriptome data were produced from 13 resistant clones and analyzed to identify secondary mutations and gene expression changes. Various mechanisms, including phenotype switching and metabolic reprogramming, have been determined to contribute to resistance development differently for each clone. The roles of microphthalmia-associated transcription factor, the master transcription factor in melanocyte differentiation/dedifferentiation, were highlighted in terms of phenotype switching. Our study provides an omics-based comprehensive overview of the molecular mechanisms governing acquired resistance to BRAF inhibitor therapy.
Collapse
Affiliation(s)
- Jiyeon Han
- Department of Bio-information Science, Ewha Womans University, Seoul 03760, Korea
| | - Yeonjoo Jung
- Ewha Research Center for Systems Biology (ERCSB), Ewha Womans University, Seoul 03760, Korea
| | - Yukyung Jun
- Ewha Research Center for Systems Biology (ERCSB), Ewha Womans University, Seoul 03760, Korea
| | - Sungsu Park
- Center for Supercomputing Application, Division of National Supercomputing, Korea Institute of Science and Technology Information, Daejeon 34141, Korea
| | - Sanghyuk Lee
- Department of Bio-information Science, Ewha Womans University, Seoul 03760, Korea
- Ewha Research Center for Systems Biology (ERCSB), Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
43
|
Peppicelli S, Andreucci E, Ruzzolini J, Bianchini F, Nediani C, Supuran CT, Calorini L. The Carbonic Anhydrase IX inhibitor SLC-0111 as emerging agent against the mesenchymal stem cell-derived pro-survival effects on melanoma cells. J Enzyme Inhib Med Chem 2021; 35:1185-1193. [PMID: 32396749 PMCID: PMC7269050 DOI: 10.1080/14756366.2020.1764549] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSC) take part to solid tumour-associated stroma and critically influence progression of malignancy. Our study represents a striking example of melanoma progression to a more malignant and resistant phenotype promoted by MSC and the possibility to contrast this diabolic liaison using CAIX inhibitors. In particular, we demonstrated that melanoma cells exposed to a MSC-conditioned medium switch to a more malignant phenotype, characterised by resistance to programmed cell death and endowed with an epithelial-to-mesenchymal transition and stem cell characteristics. These effects were reversed abrogating MSC CAIX activity using SLC-0111, a CAIX inhibitor. Moreover, the acquisition by melanoma cells of a Vemurafenib-resistant phenotype upon MSC-conditioned medium exposure was removed when MSC were treated with SLC-0111. Therefore, MSC may profoundly reprogramme melanoma cells towards a wide resistant phenotype through CAIX involvement, as the use of SLC-0111 is able to contrast the development of this highly risky adaptation for disease progression.
Collapse
Affiliation(s)
- Silvia Peppicelli
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Elena Andreucci
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Jessica Ruzzolini
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Francesca Bianchini
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Chiara Nediani
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | | | - Lido Calorini
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy.,Center of Excellence for Research, Transfer and High Education, DenoTHE University of Florence, Florence, Italy
| |
Collapse
|
44
|
Melanoma Cell Resistance to Vemurafenib Modifies Inter-Cellular Communication Signals. Biomedicines 2021; 9:biomedicines9010079. [PMID: 33467521 PMCID: PMC7830125 DOI: 10.3390/biomedicines9010079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 12/15/2022] Open
Abstract
The therapeutic success of BRAF inhibitors (BRAFi) and MEK inhibitors (MEKi) in BRAF-mutant melanoma is limited by the emergence of drug resistance, and several lines of evidence suggest that changes in the tumor microenvironment can play a pivotal role in acquired resistance. The present study focused on secretome profiling of melanoma cells sensitive or resistant to the BRAFi vemurafenib. Proteomic and cytokine/chemokine secretion analyses were performed in order to better understand the interplay between vemurafenib-resistant melanoma cells and the tumor microenvironment. We found that vemurafenib-resistant melanoma cells can influence dendritic cell (DC) maturation by modulating their activation and cytokine production. In particular, human DCs exposed to conditioned medium (CM) from vemurafenib-resistant melanoma cells produced higher levels of pro-inflammatory cytokines—that potentially facilitate melanoma growth—than DCs exposed to CM derived from parental drug-sensitive cells. Bioinformatic analysis performed on proteins identified by mass spectrometry in the culture medium from vemurafenib-sensitive and vemurafenib-resistant melanoma cells suggests a possible involvement of the proteasome pathway. Moreover, our data confirm that BRAFi-resistant cells display a more aggressive phenotype compared to parental ones, with a significantly increased production of interferon-γ, interleukin-8, vascular-endothelial growth factor, CD147/basigin, and metalloproteinase 2 (MMP-2). Plasma levels of CD147/basigin and MMP-2 were also measured before the start of therapy and at disease progression in a small group of melanoma patients treated with vemurafenib or vemurafenib plus cobimetinib. A significant increment in CD147/basigin and MMP-2 was observed in all patients at the time of treatment failure, strengthening the hypothesis that CD147/basigin might play a role in BRAFi resistance.
Collapse
|
45
|
Abstract
Since the first resection of melanoma by Hunter in 1787, efforts to treat patients with this deadly malignancy have been ongoing. Initial work to understand melanoma biology for therapeutics development began with the employment of isolated cancer cells grown in cell cultures. However, these models lack in vivo interactions with the tumor microenvironment. Melanoma cell line transplantation into suitable animals such as mice has been informative and useful for testing therapeutics as a preclinical model. Injection of freshly isolated patient melanomas into immunodeficient animals has shown the capacity to retain the genetic heterogeneity of the tumors, which is lost during the long-term culture of melanoma cells. Upon advancement of technology, genetically engineered animals have been generated to study the spontaneous development of melanomas in light of newly discovered genetic aberrations associated with melanoma formation. Culturing melanoma cells in a matrix generate tumor spheroids, providing an in vitro environment that promotes the heterogeneity commonplace with human melanoma and displaces the need for animal care facilities. Advanced 3D cultures have been created simulating the structure and cellularity of human skin to permit in vitro testing of therapeutics on melanomas expressing the same phenotype as demonstrated in vivo. This review will discuss these models and their relevance to the study of melanomagenesis, growth, metastasis, and therapy.
Collapse
Affiliation(s)
- Randal K Gregg
- Department of Basic Medical Sciences, DeBusk College of Osteopathic Medicine at Lincoln Memorial University-Knoxville, Knoxville, TN, USA.
| |
Collapse
|
46
|
Béal J, Pantolini L, Noël V, Barillot E, Calzone L. Personalized logical models to investigate cancer response to BRAF treatments in melanomas and colorectal cancers. PLoS Comput Biol 2021; 17:e1007900. [PMID: 33507915 PMCID: PMC7872233 DOI: 10.1371/journal.pcbi.1007900] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 02/09/2021] [Accepted: 12/21/2020] [Indexed: 11/19/2022] Open
Abstract
The study of response to cancer treatments has benefited greatly from the contribution of different omics data but their interpretation is sometimes difficult. Some mathematical models based on prior biological knowledge of signaling pathways facilitate this interpretation but often require fitting of their parameters using perturbation data. We propose a more qualitative mechanistic approach, based on logical formalism and on the sole mapping and interpretation of omics data, and able to recover differences in sensitivity to gene inhibition without model training. This approach is showcased by the study of BRAF inhibition in patients with melanomas and colorectal cancers who experience significant differences in sensitivity despite similar omics profiles. We first gather information from literature and build a logical model summarizing the regulatory network of the mitogen-activated protein kinase (MAPK) pathway surrounding BRAF, with factors involved in the BRAF inhibition resistance mechanisms. The relevance of this model is verified by automatically assessing that it qualitatively reproduces response or resistance behaviors identified in the literature. Data from over 100 melanoma and colorectal cancer cell lines are then used to validate the model's ability to explain differences in sensitivity. This generic model is transformed into personalized cell line-specific logical models by integrating the omics information of the cell lines as constraints of the model. The use of mutations alone allows personalized models to correlate significantly with experimental sensitivities to BRAF inhibition, both from drug and CRISPR targeting, and even better with the joint use of mutations and RNA, supporting multi-omics mechanistic models. A comparison of these untrained models with learning approaches highlights similarities in interpretation and complementarity depending on the size of the datasets. This parsimonious pipeline, which can easily be extended to other biological questions, makes it possible to explore the mechanistic causes of the response to treatment, on an individualized basis.
Collapse
Affiliation(s)
- Jonas Béal
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Lorenzo Pantolini
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Vincent Noël
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Emmanuel Barillot
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Laurence Calzone
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| |
Collapse
|
47
|
Hartman ML, Gajos-Michniewicz A, Talaj JA, Mielczarek-Lewandowska A, Czyz M. BH3 mimetics potentiate pro-apoptotic activity of encorafenib in BRAF V600E melanoma cells. Cancer Lett 2020; 499:122-136. [PMID: 33259900 DOI: 10.1016/j.canlet.2020.11.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/06/2020] [Accepted: 11/24/2020] [Indexed: 12/29/2022]
Abstract
BRAFV600- and MEK1/2-targeting therapies rarely produce durable response in melanoma patients. We investigated five BRAFV600E melanoma cell lines derived from drug-naïve tumor specimens to assess cell death response to encorafenib (Braftovi), a recently FDA-approved BRAFV600 inhibitor. Drug-naïve cell lines (i) did not harbor damaging alterations in genes encoding core apoptotic machinery, but they differed in (ii) mitochondrial priming as demonstrated by whole-cell BH3 profiling, and (iii) levels of selected anti-apoptotic proteins. Encorafenib modulated the balance between apoptosis-regulating proteins as it upregulated BIM and BMF, and attenuated NOXA, but did not affect the levels of pro-survival proteins except for MCL-1 and BCL-XL in selected cell lines. Induction of apoptosis could be predicted using Dynamic BH3 profiling. The extent of apoptosis was dependent on both (i) cell-intrinsic proximity to the apoptotic threshold (initial mitochondrial priming) and (ii) the abundance of encorafenib-induced BIM (iBIM; drug-induced change in priming). While co-inhibition of MCL-1 and BCL-XL/BCL-2 was indispensable for apoptosis in drug-naïve cells, encorafenib altered cell dependence to MCL-1, and reliance on BCL-XL/BCL-2 was additionally found in cell lines that were highly primed to apoptosis by encorafenib. This translated into robust apoptosis when encorafenib was combined with selective BH3 mimetics. Our study provides a mechanistic insight into the role of proteins from the BCL-2 family in melanoma cell response to targeted therapy, and presents preclinical evidence that (i) MCL-1 is a druggable target to potentiate encorafenib activity, whereas (ii) pharmacological inhibition of BCL-XL/BCL-2 might be relevant but only for a narrow group of encorafenib-treated patients.
Collapse
Affiliation(s)
- Mariusz L Hartman
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215, Lodz, Poland.
| | - Anna Gajos-Michniewicz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215, Lodz, Poland
| | - Julita A Talaj
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215, Lodz, Poland
| | | | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215, Lodz, Poland
| |
Collapse
|
48
|
Selective Oral MEK1/2 Inhibitor Pimasertib in Metastatic Melanoma: Antitumor Activity in a Phase I, Dose-Escalation Trial. Target Oncol 2020; 16:47-57. [PMID: 33211315 DOI: 10.1007/s11523-020-00767-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Pimasertib is a selective, potent mitogen-activated protein kinase kinase (MEK) 1/2 inhibitor. OBJECTIVES The aim of this study was to describe the efficacy, safety, and pharmacodynamics of pimasertib at pharmacologically active doses in a cohort of patients with locally advanced/metastatic melanoma from a first-in-human study of pimasertib. METHODS This was a phase I, open-label, two-part, dose-escalation study. Part 1 was conducted in patients with solid tumors and identified the maximum tolerated dose, while Part 2 was restricted to patients with advanced/metastatic melanoma. Endpoints included safety, pharmacodynamics, and antitumor activity. We present data for patients with melanoma only from both parts of the study. RESULTS In total, 93 patients with melanoma received pimasertib, 89 of whom received pharmacologically active doses (28-255 mg/day) across four dose regimens in the two parts of the study. The objective response rate was 12.4% (11/89): complete response (n = 1) and partial response (PR; n = 10). Six patients responded for > 24 weeks. Nine of the 11 responders had tumors with B-Raf Proto-Oncogene, Serine/Threonine Kinase (BRAF; n = 6) and/or NRAS Proto-Oncogene, GTPase (NRAS; n = 3) mutations. Forty-six patients had stable disease (SD). In patients with ocular melanoma (n = 13), best overall response was PR (n = 1), SD (n = 11), and disease progression (n = 1). Phosphorylated extracellular signal-regulated kinase (pERK) levels were substantially reduced within 2 h of treatment and inhibition was sustained with continuous twice-daily dosing. Treatment-related, recurrent, grade 3 or higher adverse events were reported in eight patients, including diarrhea, and skin and ocular events. CONCLUSION Results from this phase I study indicate that pimasertib has clinical activity in patients with locally advanced/metastatic melanoma, particularly BRAF- and NRAS-mutated tumors, at clinically relevant doses associated with pERK inhibition in peripheral blood mononuclear cells. TRIAL REGISTRATION ClinicalTrials.gov, NCT00982865.
Collapse
|
49
|
Aoude LG, Bonazzi VF, Brosda S, Patel K, Koufariotis LT, Oey H, Nones K, Wood S, Pearson JV, Lonie JM, Arneil M, Atkinson V, Smithers BM, Waddell N, Barbour AP. Pathogenic germline variants are associated with poor survival in stage III/IV melanoma patients. Sci Rep 2020; 10:17687. [PMID: 33077847 PMCID: PMC7572377 DOI: 10.1038/s41598-020-74956-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Patients with late stage resected cutaneous melanoma have poor overall survival (OS) and experience irreversible adverse events from systemic therapy. There is a clinical need to identify biomarkers to predict outcome. Performing germline/tumour whole-exome sequencing of 44 stage III/IV melanoma patients we identified pathogenic germline mutations in CDKN2A, CDK4, ATM, POLH, MRE11A, RECQL4 and XPC, affecting 7/44 patients. These mutations were associated with poor OS (p = 0.0082). We confirmed our findings in The Cancer Genome Atlas (TCGA) human skin cutaneous melanoma cohort where we identified pathogenic variants in 40/455 patients (p = 0.0203). Combining these cohorts (n = 499) further strengthened these findings showing germline carriers had worse OS (p = 0.0009). Additionally, we determined whether tumour mutation burden (TMB) or BRAF status were prognostic markers of survival. Low TMB rate (< 20 Mut/Mb; p = 0.0034) and BRAF p.V600 mutation (p = 0.0355) were associated with worse progression-free survival. Combining these biomarkers indicated that V600 mutant patients had significantly lower TMB (p = 0.0155). This was confirmed in the TCGA (n = 443, p = 0.0007). Integrative analysis showed germline mutation status conferred the highest risk (HR 5.2, 95% CI 1.72–15.7). Stage IV (HR 2.5, 0.74–8.6) and low TMB (HR 2.3, 0.57–9.4) were similar, whereas BRAF V600 status was the weakest prognostic biomarker (HR 1.5, 95% CI 0.44–5.2).
Collapse
Affiliation(s)
- Lauren G Aoude
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| | - Vanessa F Bonazzi
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Sandra Brosda
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Kalpana Patel
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | | | - Harald Oey
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Katia Nones
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Scott Wood
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - John V Pearson
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - James M Lonie
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Melissa Arneil
- Division of Cancer Services, Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia
| | - Victoria Atkinson
- Queensland Melanoma Project, Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia.,Faculty of Medicine, University of Queensland, St Lucia, QLD, 4067, Australia
| | - B Mark Smithers
- Queensland Melanoma Project, Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia.,Faculty of Medicine, University of Queensland, St Lucia, QLD, 4067, Australia
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Andrew P Barbour
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia.,Queensland Melanoma Project, Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia
| |
Collapse
|
50
|
Horizontal Combination of MEK and PI3K/mTOR Inhibition in BRAF Mutant Tumor Cells with or without Concomitant PI3K Pathway Mutations. Int J Mol Sci 2020; 21:ijms21207649. [PMID: 33081092 PMCID: PMC7589607 DOI: 10.3390/ijms21207649] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/22/2020] [Accepted: 10/14/2020] [Indexed: 12/19/2022] Open
Abstract
The RAS/RAF and PI3K/Akt pathways play a key regulatory role in cancer and are often hit by oncogenic mutations. Despite molecular targeting, the long-term success of monotherapy is often hampered by de novo or acquired resistance. In the case of concurrent mutations in both pathways, horizontal combination could be a reasonable approach. In our study, we investigated the MEK inhibitor selumetinib and PI3K/mTOR dual inhibitor BEZ235 alone and in combination in BRAF-only mutant and BRAF + PI3K/PTEN double mutant cancer cells using short- and long-term 2D viability assays, spheroid assays, and immunoblots. In the 2D assays, selumetinib was more effective on BRAF-only mutant lines when compared to BRAF + PI3K/PTEN double mutants. Furthermore, combination therapy had an additive effect in most of the lines while synergism was observed in two of the double mutants. Importantly, in the SW1417 BRAF + PI3K double mutant cells, synergism was also confirmed in the spheroid and in the in vivo model. Mechanistically, p-Akt level decreased only in the SW1417 cell line after combination treatment. In conclusion, the presence of concurrent mutations alone did not predict a stronger response to combination treatment. Therefore, additional investigations are warranted to identify predictive factors that can select patients who can benefit from the horizontal combinational inhibition of these two pathways.
Collapse
|