1
|
Hicks HM, Nassar VL, Lund J, Rose MM, Schweppe RE. The effects of Aurora Kinase inhibition on thyroid cancer growth and sensitivity to MAPK-directed therapies. Cancer Biol Ther 2024; 25:2332000. [PMID: 38521968 PMCID: PMC10962586 DOI: 10.1080/15384047.2024.2332000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 03/14/2024] [Indexed: 03/25/2024] Open
Abstract
Thyroid cancer is one of the deadliest endocrine cancers, and its incidence has been increasing. While mutations in BRAF are common in thyroid cancer, advanced PTC patients currently lack therapeutic options targeting the MAPK pathway, and despite the approved combination of BRAF and MEK1/2 inhibition for BRAF-mutant ATC, resistance often occurs. Here, we assess growth and signaling responses to combined BRAF and MEK1/2 inhibition in a panel of BRAF-mutant thyroid cancer cell lines. We first showed that combined BRAF and MEK1/2 inhibition synergistically inhibits cell growth in four out of six of the -BRAF-mutant thyroid cancer cell lines tested. Western blotting showed that the MAPK pathway was robustly inhibited in all cell lines. Therefore, to identify potential mechanisms of resistance, we performed RNA-sequencing in cells sensitive or resistant to MEK1/2 inhibition. In response to MEK1/2 inhibition, we identified a downregulation of Aurora Kinase B (AURKB) in sensitive but not resistant cells. We further demonstrated that combined MEK1/2 and AURKB inhibition slowed cell growth, which was phenocopied by inhibiting AURKB and ERK1/2. Finally, we show that combined AURKB and ERK1/2 inhibition induces apoptosis in BRAF-mutant thyroid cancer cell lines, together suggesting a potential combination therapy for BRAF-mutant thyroid cancer patients.
Collapse
Affiliation(s)
- Hannah M. Hicks
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Veronica L. Nassar
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jane Lund
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Madison M. Rose
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Rebecca E. Schweppe
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
2
|
Gutierrez JJG, Lau E, Dharmapalan S, Parker M, Chen Y, Álvarez MA, Wang D. Multi-output prediction of dose-response curves enables drug repositioning and biomarker discovery. NPJ Precis Oncol 2024; 8:209. [PMID: 39304771 DOI: 10.1038/s41698-024-00691-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 08/28/2024] [Indexed: 09/22/2024] Open
Abstract
Drug response prediction is hampered by uncertainty in the measures of response and selection of doses. In this study, we propose a probabilistic multi-output model to simultaneously predict all dose-responses and uncover their biomarkers. By describing the relationship between genomic features and chemical properties to every response at every dose, our multi-output Gaussian Process (MOGP) models enable assessment of drug efficacy using any dose-response metric. This approach was tested across two drug screening studies and ten cancer types. Kullback-leibler divergence measured the importance of each feature and identified EZH2 gene as a novel biomarker of BRAF inhibitor response. We demonstrate the effectiveness of our MOGP models in accurately predicting dose-responses in different cancer types and when there is a limited number of drug screening experiments for training. Our findings highlight the potential of MOGP models in enhancing drug development pipelines by reducing data requirements and improving precision in dose-response predictions.
Collapse
Affiliation(s)
- Juan-José Giraldo Gutierrez
- National Heart and Lung Institute, Imperial College London, London, UK.
- Department of Computer Science, The University of Sheffield, Sheffield, UK.
| | - Evelyn Lau
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Subhashini Dharmapalan
- Department of Computer Science, The University of Sheffield, Sheffield, UK
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Melody Parker
- Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Yurui Chen
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
- Department of Mathematics, National University of Singapore, Singapore, Republic of Singapore
| | - Mauricio A Álvarez
- Department of Computer Science, The University of Manchester, Manchester, UK
| | - Dennis Wang
- National Heart and Lung Institute, Imperial College London, London, UK.
- Department of Computer Science, The University of Sheffield, Sheffield, UK.
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore.
- Bioinformatics Institute (BII), Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore.
| |
Collapse
|
3
|
Roccuzzo G, Fava P, Astrua C, Brizio MG, Cavaliere G, Bongiovanni E, Santaniello U, Carpentieri G, Cangiolosi L, Brondino C, Pala V, Ribero S, Quaglino P. Real-Life Outcomes of Adjuvant Targeted Therapy and Anti-PD1 Agents in Stage III/IV Resected Melanoma. Cancers (Basel) 2024; 16:3095. [PMID: 39272953 PMCID: PMC11394626 DOI: 10.3390/cancers16173095] [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: 07/29/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024] Open
Abstract
This study was carried out at the Dermatologic Clinic of the University of Turin, Italy, to assess the effectiveness and safety of adjuvant therapy in patients who received either targeted therapy (TT: dabrafenib + trametinib) or immunotherapy (IT: nivolumab or pembrolizumab) for up to 12 months. A total of 163 patients participated, including 147 with stage III and 19 with stage IV with no evidence of disease. The primary outcomes were relapse-free survival (RFS), distant metastasis-free survival (DMFS), and overall survival (OS). At 48 months, both TT and IT approaches yielded comparable outcomes in terms of RFS (55.6-55.4%, p = 0.532), DMFS (58.2-59.8%, p = 0.761), and OS (62.4-69.5%, p = 0.889). Whilst temporary therapy suspension was more common among TT-treated patients compared to IT-treated individuals, therapy discontinuation due to adverse events occurred at comparable rates in both groups. Predictors of relapse included mitoses, lymphovascular invasion, ulceration, and positive sentinel lymph nodes. Overall, the proportion of BRAF-mutated patients receiving IT stood at 7.4%, lower than what was observed in clinical trials.
Collapse
Affiliation(s)
- Gabriele Roccuzzo
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy
| | - Paolo Fava
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy
| | - Chiara Astrua
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy
| | - Matteo Giovanni Brizio
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy
| | - Giovanni Cavaliere
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy
| | - Eleonora Bongiovanni
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy
| | - Umberto Santaniello
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy
| | - Giulia Carpentieri
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy
| | - Luca Cangiolosi
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy
| | - Camilla Brondino
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy
| | - Valentina Pala
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy
| | - Simone Ribero
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy
| | - Pietro Quaglino
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy
| |
Collapse
|
4
|
Luo Y, Su B, Hung V, Luo Y, Shi Y, Wang G, de Graaf D, Dinarello CA, Spaner DE. IL-1 receptor antagonism reveals a yin-yang relationship between NFκB and interferon signaling in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A 2024; 121:e2405644121. [PMID: 39121163 PMCID: PMC11331101 DOI: 10.1073/pnas.2405644121] [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: 03/26/2024] [Accepted: 07/10/2024] [Indexed: 08/11/2024] Open
Abstract
Nuclear factor kappa B (NFκB) is a pathogenic factor in chronic lymphocytic leukemia (CLL) that is not addressed specifically by current therapies. NFκB is activated by inflammatory factors that stimulate toll-like receptors (TLRs) and receptors for interleukin-1 (IL-1) family members. IL-1 is considered a master regulator of inflammation, and IL-1 receptor signaling is inhibited by the IL-1 receptor antagonist anakinra. These considerations suggested that anakinra might have a role in the treatment of CLL. Consistent with this idea, anakinra inhibited spontaneous and TLR7-mediated activation of the canonical NFκB pathway in CLL cells in vitro. However, CLL cells exhibited only weak signaling responses to IL-1 itself, and anakinra was found to inhibit NFκB along with oxidative stress in an IL-1 receptor-independent manner. Anakinra was then administered with minimal toxicity to 11 previously untreated CLL patients in a phase I dose-escalation trial (NCT04691765). A stereotyped clinical response was observed in all patients. Anakinra lowered blood lymphocytes and lymph node sizes within the first month that were associated with downregulation of NFκB and oxidative stress in the leukemia cells. However, inhibition of NFκB was accompanied by upregulation of type 1 interferon (IFN) signaling, c-MYC-regulated genes and proteins, and loss of the initial clinical response. Anakinra increased IFN signaling and survival of CLL cells in vitro that were, respectively, phenocopied by mitochondrial antioxidants and reversed by IFN receptor blocking antibodies. These observations suggest that anakinra has activity in CLL and may be a useful adjunct for conventional therapies as long as compensatory IFN signaling is blocked at the same time.
Collapse
Affiliation(s)
- YuXuan Luo
- Biological Science Platform, Sunnybrook Research Institute, Sunnybrook hospital, Toronto M4N 3M5, Canada
- Department of Immunology, University of Toronto, Toronto M5S 1A8, Canada
| | - BoYang Su
- Biological Science Platform, Sunnybrook Research Institute, Sunnybrook hospital, Toronto M4N 3M5, Canada
- Department of Medical Biophysics, University of Toronto, Toronto M5G 2M9, Canada
| | - Vincent Hung
- Biological Science Platform, Sunnybrook Research Institute, Sunnybrook hospital, Toronto M4N 3M5, Canada
| | - YuHan Luo
- Biological Science Platform, Sunnybrook Research Institute, Sunnybrook hospital, Toronto M4N 3M5, Canada
- Department of Immunology, University of Toronto, Toronto M5S 1A8, Canada
| | - Yonghong Shi
- Biological Science Platform, Sunnybrook Research Institute, Sunnybrook hospital, Toronto M4N 3M5, Canada
| | - Guizhi Wang
- Biological Science Platform, Sunnybrook Research Institute, Sunnybrook hospital, Toronto M4N 3M5, Canada
| | - Dennis de Graaf
- Department of Medicine, University of Colorado Denver, Denver, CO 80045
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn 53127, Germany
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Denver, CO 80045
- Department of Medicine, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands
| | - David E Spaner
- Biological Science Platform, Sunnybrook Research Institute, Sunnybrook hospital, Toronto M4N 3M5, Canada
- Department of Immunology, University of Toronto, Toronto M5S 1A8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto M5G 2M9, Canada
- Department of Hematology, Odette Cancer Center, Toronto M4N 3M5, Canada
- Department of Medicine, University of Toronto, Toronto M5G 2C4, Canada
| |
Collapse
|
5
|
Luan W, Lu X, Peng H, Shen X, Rao M, Ruan H. Exosomal miR-19a derived from melanoma cell promotes the vemurafenib resistance of malignant melanoma through directly targeting LRIG1 to reactivate AKT and MAPK pathway. Pathol Res Pract 2024; 260:155410. [PMID: 38955119 DOI: 10.1016/j.prp.2024.155410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 04/04/2024] [Accepted: 06/13/2024] [Indexed: 07/04/2024]
Abstract
Exosomes derived from neighboring v-raf murine sarcoma viral oncogene homolog B1 inhibitor (BRAFi)-resistant melanoma cells mediate the formation of resistance in melanoma cells sensitive to BRAFi. The function and molecular mechanisms of exosomal miRNA in BRAFi resistance of melanoma have not been studied. We found that the expression of miR-19a in BRAFi resistant melanoma cells was significantly higher than that in sensitive cells, and miR-19a contributes to the resistance of melanoma cells to BRAFi by targeting immunoglobulin-like domains protein 1 (LRIG1). miR-19a was highly enriched in exosomes secreted from BRAFi resistant melanoma cells, and these exosomal miR-19a promote the spread of BRAFi resistant. The reactivation of Protein kinase B (AKT) and mitogen-activated protein kinase (MAPK) pathways is the main reason for the BRAFi resistant of melanoma cells. We demonstrated that exosomal miR-19a derived from melanoma cell promotes the formation and spread of BRAFi resistant in melanoma through targeting LRIG1 to reactivate AKT and MAPK pathway. Therefore, miR-19a may serve as a potential therapeutic target in melanoma patients with acquired drug resistance.
Collapse
Affiliation(s)
- Wenkang Luan
- Department of Plastic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xu Lu
- Department of Plastic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Huiyong Peng
- Department of Laboratory Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang Medical School of Nanjing Medical University, Zhenjiang, Jiangsu, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuanlin Shen
- Department of Rehabilitation, Changshu No. 2 People's Hospital (Changshu Hospital affiliated the NanTong University), Changshu, Jiangsu 215500, China
| | - Min Rao
- Hepatobiliary surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Hongru Ruan
- Department of Plastic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
6
|
Kim KB. Personalized therapy in oncology: melanoma as a paradigm for molecular-targeted treatment approaches. Clin Exp Metastasis 2024; 41:465-471. [PMID: 38935186 DOI: 10.1007/s10585-024-10291-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 04/29/2024] [Indexed: 06/28/2024]
Abstract
In recent decades, the field of systemic cancer treatment has seen remarkable changes due to advancements in the understanding of cancer's biology, immunology, and genetic makeup. As a result, individuals with late-stage cancers are now achieving survival rates that were previously unattainable. The goal of personalized cancer therapy is to enhance clinical outcomes by customizing drug treatments to suit the unique genetic and/or epigenetic profiles of each patient's tumor. This approach aims to reduce the side effects commonly associated with ineffective treatments. Advances in genetic sequencing and molecular cytogenetics have been instrumental in identifying cancer-driving mutations and epigenetic irregularities, leading to the development of specific molecular therapies. This review article highlights the progress and success of targeted molecular therapies in treating malignant melanoma, illustrating the concept of personalized cancer treatment.
Collapse
Affiliation(s)
- Kevin B Kim
- California Pacific Medical Center Research Institute, 2333 Buchanan St, San Francisco, CA, 94115, USA.
| |
Collapse
|
7
|
Featherston T, Paumann-Page M, Hampton MB. Melanoma redox biology and the emergence of drug resistance. Adv Cancer Res 2024; 162:145-171. [PMID: 39069368 DOI: 10.1016/bs.acr.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Melanoma is the deadliest form of skin cancer, with the loss of approximately 60,000 lives world-wide each year. Despite the development of targeted therapeutics, including compounds that have selectivity for mutant oncoproteins expressed only in cancer cells, many patients are either unresponsive to initial therapy or their tumors acquire resistance. This results in five-year survival rates of below 25%. New strategies that either kill drug-resistant melanoma cells or prevent their emergence would be extremely valuable. Melanoma, like other cancers, has long been described as being under increased oxidative stress, resulting in an increased reliance on antioxidant defense systems. Changes in redox homeostasis are most apparent during metastasis and during the metabolic reprogramming associated with the development of treatment resistance. This review discusses oxidative stress in melanoma, with a particular focus on targeting antioxidant pathways to limit the emergence of drug resistant cells.
Collapse
Affiliation(s)
- Therese Featherston
- Mātai Hāora-Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Martina Paumann-Page
- Mātai Hāora-Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.
| | - Mark B Hampton
- Mātai Hāora-Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
8
|
Boutalaka M, El Bahi S, Alaqarbeh M, El Alaouy MA, Koubi Y, Khatabi KE, Maghat H, Bouachrine M, Lakhlifi T. Computational investigation of imidazo[2,1-b]oxazole derivatives as potential mutant BRAF kinase inhibitors: 3D-QSAR, molecular docking, molecular dynamics simulation, and ADMETox studies. J Biomol Struct Dyn 2024; 42:5268-5287. [PMID: 37424193 DOI: 10.1080/07391102.2023.2233629] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/09/2023] [Indexed: 07/11/2023]
Abstract
BRAF inhibitors are known to be an effective therapeutic target for treating melanoma and other types of cancer. Using 3D-QSAR, molecular docking, and MD simulations, this study evaluated various imidazo[2,1-b]oxazole derivatives that function as mutant BRAF kinase inhibitors. Comparative molecular field analysis (CoMFA) and comparative molecular similarity index analysis (CoMSIA) were used to create the 3D-QSAR models. CoMSIA/SEHA model has solid predictive power across several models (Q2 = 0.578; R2 = 0.828; R2pred = 0.74) and is the best model according to the numerous field models generated. The created model's predictive power was evaluated through external validation using a test set. CoMSIA/SEHA contour maps collect information that can be used to identify critical regions with solid anticancer activity. We developed four inhibitors with high predicted activity due to these observations. ADMET prediction was used to assess the toxicity of the proposed imidazo[2,1-b]oxazole compounds. The predictive molecules (T1-T4) demonstrated good ADMET properties, excluding the toxic active compounds 11r from the database. Molecular docking was also used to determine the patterns and modes of interactions between imidazo[2,1-b]oxazole ligands and receptors, which revealed that the proposed imidazo[2,1-b]oxazole scaffold was stable in the receptor's active site (PDB code: 4G9C). The suggested compounds (T1-T4) were subjected to molecular dynamics simulations lasting 100 ns to determine their binding free energies. The results showed that T2 had a more favorable binding free energy (-149.552 kJ/mol) than T1 (-112.556 kJ/mol), T3 (-115.503 kJ/mol), and T4 (-102.553 kJ/mol). The results suggest that the imidazo[2,1-b]oxazole compounds investigated in this study have potential as inhibitors of BRAF kinase and could be further developed as anticancer drugs. Highlights22 imidazo[2,1-b]oxazole compounds were subjected to research on three-dimensional quantitative conformational relationships.Using contour maps from 3D-QSAR models as a guide was used to figure out the areas and strategies for structural optimization.Combined molecular docking, molecular dynamics simulations, and binding free energy calculations to verify the inhibitor activity of the proposed 22 imidazo[2,1-b]oxazole compounds.Four potential B-RAF Kinase inhibitors were discovered, providing theoretical clues for developing a highly anticancer agent.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Meryem Boutalaka
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
| | - Salma El Bahi
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
| | | | - Moulay Ahfid El Alaouy
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
| | - Yassine Koubi
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
| | - Khalil El Khatabi
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
| | - Hamid Maghat
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
| | - Mohammed Bouachrine
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
- EST Khenifra, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Tahar Lakhlifi
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
| |
Collapse
|
9
|
Tanese K, Ogata D. The role of macrophage migration inhibitory factor family and CD74 in the pathogenesis of melanoma. Exp Dermatol 2024; 33:e15122. [PMID: 38884501 DOI: 10.1111/exd.15122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/18/2024]
Abstract
Melanoma is an aggressive tumour with poor prognosis that arises from the malignant transformation of melanocytes. Over the past few decades, intense research into the pathogenesis of melanoma has led to the development of BRAF and immune checkpoint inhibitors, including antibodies against programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4), which have shown clinically significant efficacy. However, some tumours do not respond to these therapies initially or become treatment resistant. Most melanoma tissues appear to possess biological characteristics that allow them to evade these treatments, and identifying these characteristics is one of the major challenges facing cancer researchers. One such characteristic that has recently gained attention is the role of macrophage migration inhibitory factor (MIF) and its receptor CD74. This review outlines the cellular and molecular functions of CD74, MIF and their family of proteins. We then review their roles in tumours based on previous reports, highlight their pathological significance in melanoma and discuss their potential as therapeutic targets.
Collapse
Affiliation(s)
- Keiji Tanese
- Department of Dermatology, Toho University School of Medicine, Tokyo, Japan
| | - Dai Ogata
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
10
|
De Carli A, Kapelyukh Y, Kursawe J, Chaplain MAJ, Wolf CR, Hamis S. Simulating BRAFV600E-MEK-ERK signalling dynamics in response to vertical inhibition treatment strategies. NPJ Syst Biol Appl 2024; 10:51. [PMID: 38750040 PMCID: PMC11096323 DOI: 10.1038/s41540-024-00379-9] [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: 12/13/2023] [Accepted: 04/29/2024] [Indexed: 05/18/2024] Open
Abstract
In vertical inhibition treatment strategies, multiple components of an intracellular pathway are simultaneously inhibited. Vertical inhibition of the BRAFV600E-MEK-ERK signalling pathway is a standard of care for treating BRAFV600E-mutated melanoma where two targeted cancer drugs, a BRAFV600E-inhibitor, and a MEK inhibitor, are administered in combination. Targeted therapies have been linked to early onsets of drug resistance, and thus treatment strategies of higher complexities and lower doses have been proposed as alternatives to current clinical strategies. However, finding optimal complex, low-dose treatment strategies is a challenge, as it is possible to design more treatment strategies than are feasibly testable in experimental settings. To quantitatively address this challenge, we develop a mathematical model of BRAFV600E-MEK-ERK signalling dynamics in response to combinations of the BRAFV600E-inhibitor dabrafenib (DBF), the MEK inhibitor trametinib (TMT), and the ERK-inhibitor SCH772984 (SCH). From a model of the BRAFV600E-MEK-ERK pathway, and a set of molecular-level drug-protein interactions, we extract a system of chemical reactions that is parameterised by in vitro data and converted to a system of ordinary differential equations (ODEs) using the law of mass action. The ODEs are solved numerically to produce simulations of how pathway-component concentrations change over time in response to different treatment strategies, i.e., inhibitor combinations and doses. The model can thus be used to limit the search space for effective treatment strategies that target the BRAFV600E-MEK-ERK pathway and warrant further experimental investigation. The results demonstrate that DBF and DBF-TMT-SCH therapies show marked sensitivity to BRAFV600E concentrations in silico, whilst TMT and SCH monotherapies do not.
Collapse
Affiliation(s)
- Alice De Carli
- School of Mathematics and Statistics, University of St Andrews, St Andrews, Scotland, UK
| | - Yury Kapelyukh
- School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, UK
| | - Jochen Kursawe
- School of Mathematics and Statistics, University of St Andrews, St Andrews, Scotland, UK
| | - Mark A J Chaplain
- School of Mathematics and Statistics, University of St Andrews, St Andrews, Scotland, UK
| | - C Roland Wolf
- School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, UK
| | - Sara Hamis
- School of Mathematics and Statistics, University of St Andrews, St Andrews, Scotland, UK.
- Tampere Institute for Advanced Study, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Department of Information Technology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
11
|
Algazi AP, Moon J, Lao CD, Chmielowski B, Kendra KL, Lewis KD, Gonzalez R, Kim K, Godwin JE, Curti BD, Latkovic-Taber M, Lomeli SH, Gufford BT, Scumpia PO, Lo RS, Othus M, Ribas A. A phase 1 study of triple-targeted therapy with BRAF, MEK, and AKT inhibitors for patients with BRAF-mutated cancers. Cancer 2024; 130:1784-1796. [PMID: 38261444 DOI: 10.1002/cncr.35200] [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] [Revised: 10/30/2023] [Accepted: 11/21/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND Aberrant PI3K/AKT signaling in BRAF-mutant cancers contributes to resistance to BRAF inhibitors. The authors examined dual MAPK and PI3K pathway inhibition in patients who had BRAF-mutated solid tumors (ClinicalTrials.gov identifier NCT01902173). METHODS Patients with BRAF V600E/V600K-mutant solid tumors received oral dabrafenib at 150 mg twice daily with dose escalation of oral uprosertib starting at 50 mg daily, or, in the triplet cohorts, with dose escalation of both oral trametinib starting at 1.5 mg daily and oral uprosertib starting at 25 mg daily. Dose-limiting toxicities (DLTs) were assessed within the first 56 days of treatment. Radiographic responses were assessed at 8-week intervals. RESULTS Twenty-seven patients (22 evaluable) were enrolled in parallel doublet and triplet cohorts. No DLTs were observed in the doublet cohorts (N = 7). One patient had a DLT at the maximum administered dose of triplet therapy (dabrafenib 150 mg twice daily and trametinib 2 mg daily plus uprosertib 75 mg daily). Three patients in the doublet cohorts had partial responses (including one who had BRAF inhibitor-resistant melanoma). Two patients in the triplet cohorts had a partial response, and one patient had an unconfirmed partial response. Pharmacokinetic data suggested reduced dabrafenib and dabrafenib metabolite exposure in patients who were also exposed to both trametinib and uprosertib, but not in whose who were exposed to uprosertib without trametinib. CONCLUSIONS Concomitant inhibition of both the MAPK and PI3K-AKT pathways for the treatment of BRAF-mutated cancers was well tolerated, leading to objective responses, but higher level drug-drug interactions affected exposure to dabrafenib and its metabolites.
Collapse
Affiliation(s)
- Alain P Algazi
- University of California-San Francisco, San Francisco, California, USA
| | - James Moon
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Southwest Oncology Group Statistical Center, Seattle, Washington, USA
| | | | - Bartosz Chmielowski
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, California, USA
| | - Kari L Kendra
- The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Karl D Lewis
- University of Colorado Comprehensive Cancer Center, Denver, Colorado, USA
| | - Rene Gonzalez
- University of Colorado Comprehensive Cancer Center, Denver, Colorado, USA
| | - Kevin Kim
- California Pacific Medical Center Research Institute, San Francisco, California, USA
| | | | | | | | - Shirley H Lomeli
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, California, USA
| | | | - Philip O Scumpia
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, California, USA
| | - Roger S Lo
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, California, USA
| | - Megan Othus
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Antoni Ribas
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, California, USA
| |
Collapse
|
12
|
Grogan L, Shapiro P. Progress in the development of ERK1/2 inhibitors for treating cancer and other diseases. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 100:181-207. [PMID: 39034052 DOI: 10.1016/bs.apha.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
The extracellular signal-regulated kinases-1 and 2 (ERK1/2) are ubiquitous regulators of many cellular functions, including proliferation, differentiation, migration, and cell death. ERK1/2 regulate cell functions by phosphorylating a diverse collection of protein substrates consisting of other kinases, transcription factors, structural proteins, and other regulatory proteins. ERK1/2 regulation of cell functions is tightly regulated through the balance between activating phosphorylation by upstream kinases and inactivating dephosphorylation by phosphatases. Disruption of homeostatic ERK1/2 regulation caused by elevated extracellular signals or mutations in upstream regulatory proteins leads to the constitutive activation of ERK1/2 signaling and uncontrolled cell proliferation observed in many types of cancer. Many inhibitors of upstream kinase regulators of ERK1/2 have been developed and are part of targeted therapeutic options to treat a variety of cancers. However, the efficacy of these drugs in providing sustained patient responses is limited by the development of acquired resistance often involving re-activation of ERK1/2. As such, recent drug discovery efforts have focused on the direct targeting of ERK1/2. Several ATP competitive ERK1/2 inhibitors have been identified and are being tested in cancer clinical trials. One drug, Ulixertinib (BVD-523), has received FDA approval for use in the Expanded Access Program for patients with no other therapeutic options. This review provides an update on ERK1/2 inhibitors in clinical trials, their successes and limitations, and new academic drug discovery efforts to modulate ERK1/2 signaling for treating cancer and other diseases.
Collapse
Affiliation(s)
- Lena Grogan
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Paul Shapiro
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States.
| |
Collapse
|
13
|
Fateeva A, Eddy K, Chen S. Current State of Melanoma Therapy and Next Steps: Battling Therapeutic Resistance. Cancers (Basel) 2024; 16:1571. [PMID: 38672652 PMCID: PMC11049326 DOI: 10.3390/cancers16081571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Melanoma is the most aggressive and deadly form of skin cancer due to its high propensity to metastasize to distant organs. Significant progress has been made in the last few decades in melanoma therapeutics, most notably in targeted therapy and immunotherapy. These approaches have greatly improved treatment response outcomes; however, they remain limited in their abilities to hinder disease progression due, in part, to the onset of acquired resistance. In parallel, intrinsic resistance to therapy remains an issue to be resolved. In this review, we summarize currently available therapeutic options for melanoma treatment and focus on possible mechanisms that drive therapeutic resistance. A better understanding of therapy resistance will provide improved rational strategies to overcome these obstacles.
Collapse
Affiliation(s)
- Anna Fateeva
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA; (A.F.); (K.E.)
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ 08854, USA
| | - Kevinn Eddy
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA; (A.F.); (K.E.)
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ 08854, USA
| | - Suzie Chen
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA; (A.F.); (K.E.)
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ 08854, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- U.S. Department of Veterans Affairs, New Jersey Health Care System, East Orange, NJ 07018, USA
| |
Collapse
|
14
|
Dinter L, Karitzky PC, Schulz A, Wurm AA, Mehnert MC, Sergon M, Tunger A, Lesche M, Wehner R, Müller A, Käubler T, Niessner H, Dahl A, Beissert S, Schmitz M, Meier F, Seliger B, Westphal D. BRAF and MEK inhibitor combinations induce potent molecular and immunological effects in NRAS-mutant melanoma cells: Insights into mode of action and resistance mechanisms. Int J Cancer 2024; 154:1057-1072. [PMID: 38078628 DOI: 10.1002/ijc.34807] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 10/17/2023] [Accepted: 11/03/2023] [Indexed: 01/23/2024]
Abstract
About 25% of melanoma harbor activating NRAS mutations, which are associated with aggressive disease therefore requiring a rapid antitumor intervention. However, no efficient targeted therapy options are currently available for patients with NRAS-mutant melanoma. MEK inhibitors (MEKi) appear to display a moderate antitumor activity and also immunological effects in NRAS-mutant melanoma, providing an ideal backbone for combination treatments. In our study, the MEKi binimetinib, cobimetinib and trametinib combined with the BRAF inhibitors (BRAFi) encorafenib, vemurafenib and dabrafenib were investigated for their ability to inhibit proliferation, induce apoptosis and alter the expression of immune modulatory molecules in sensitive NRAS-mutant melanoma cells using two- and three-dimensional cell culture models as well as RNA sequencing analyses. Furthermore, NRAS-mutant melanoma cells resistant to the three BRAFi/MEKi combinations were established to characterize the mechanisms contributing to their resistance. All BRAFi induced a stress response in the sensitive NRAS-mutant melanoma cells thereby significantly enhancing the antiproliferative and proapoptotic activity of the MEKi analyzed. Furthermore, BRAFi/MEKi combinations upregulated immune relevant molecules, such as ICOS-L, components of antigen-presenting machinery and the "don't eat me signal" molecule CD47 in the melanoma cells. The BRAFi/MEKi-resistant, NRAS-mutant melanoma cells counteracted the molecular and immunological effects of BRAFi/MEKi by upregulating downstream mitogen-activated protein kinase pathway molecules, inhibiting apoptosis and promoting immune escape mechanisms. Together, our study reveals potent molecular and immunological effects of BRAFi/MEKi in sensitive NRAS-mutant melanoma cells that may be exploited in new combinational treatment strategies for patients with NRAS-mutant melanoma.
Collapse
Affiliation(s)
- Lisa Dinter
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Paula C Karitzky
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
| | - Alexander Schulz
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Alexander A Wurm
- National Center for Tumor Diseases (NCT) Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- Department of Translational Medical Oncology, NCT Dresden, Dresden, Germany
- Mildred Scheel Early Career Center, NCT Dresden, Medical Faculty and University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
| | - Marie-Christin Mehnert
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Mildred Sergon
- Institute of Pathology, University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
| | - Antje Tunger
- National Center for Tumor Diseases (NCT) Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Mathias Lesche
- DRESDEN-Concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, Dresden, Germany
| | - Rebekka Wehner
- National Center for Tumor Diseases (NCT) Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anja Müller
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Theresa Käubler
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
| | - Heike Niessner
- Department of Dermatology, Oncology, University Medical Center, Tübingen, Germany
| | - Andreas Dahl
- DRESDEN-Concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, Dresden, Germany
| | - Stefan Beissert
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Marc Schmitz
- National Center for Tumor Diseases (NCT) Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- Skin Cancer Center at the University Cancer Center Dresden, University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
| | - Barbara Seliger
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Institute of Translational Immunology, Medical School "Theodor Fontane", Brandenburg an der Havel, Germany
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Dana Westphal
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| |
Collapse
|
15
|
Duan XP, Qin BD, Jiao XD, Liu K, Wang Z, Zang YS. New clinical trial design in precision medicine: discovery, development and direction. Signal Transduct Target Ther 2024; 9:57. [PMID: 38438349 PMCID: PMC10912713 DOI: 10.1038/s41392-024-01760-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 03/06/2024] Open
Abstract
In the era of precision medicine, it has been increasingly recognized that individuals with a certain disease are complex and different from each other. Due to the underestimation of the significant heterogeneity across participants in traditional "one-size-fits-all" trials, patient-centered trials that could provide optimal therapy customization to individuals with specific biomarkers were developed including the basket, umbrella, and platform trial designs under the master protocol framework. In recent years, the successive FDA approval of indications based on biomarker-guided master protocol designs has demonstrated that these new clinical trials are ushering in tremendous opportunities. Despite the rapid increase in the number of basket, umbrella, and platform trials, the current clinical and research understanding of these new trial designs, as compared with traditional trial designs, remains limited. The majority of the research focuses on methodologies, and there is a lack of in-depth insight concerning the underlying biological logic of these new clinical trial designs. Therefore, we provide this comprehensive review of the discovery and development of basket, umbrella, and platform trials and their underlying logic from the perspective of precision medicine. Meanwhile, we discuss future directions on the potential development of these new clinical design in view of the "Precision Pro", "Dynamic Precision", and "Intelligent Precision". This review would assist trial-related researchers to enhance the innovation and feasibility of clinical trial designs by expounding the underlying logic, which be essential to accelerate the progression of precision medicine.
Collapse
Affiliation(s)
- Xiao-Peng Duan
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bao-Dong Qin
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiao-Dong Jiao
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ke Liu
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhan Wang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yuan-Sheng Zang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
16
|
Guo X, Huang S, Zhang Y, Wang H, Li L, Ran J, Chen D, Li X, Li J. Evodiamine inhibits growth of vemurafenib drug-resistant melanoma via suppressing IRS4/PI3K/AKT signaling pathway. J Nat Med 2024; 78:342-354. [PMID: 38324123 DOI: 10.1007/s11418-023-01769-9] [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: 09/20/2023] [Accepted: 11/28/2023] [Indexed: 02/08/2024]
Abstract
Evodiamine, a novel alkaloid, was isolated from the fruit of tetradium. It exerts a diversity of pharmacological effects and has been used to treat gastropathy, hypertension, and eczema. Several studies reported that evodiamine has various biological effects, including anti-nociceptive, anti-bacterial, anti-obesity, and anti-cancer activities. However, there is no research regarding its effects on drug-resistant cancer. This study aimed to investigate the effect of evodiamine on human vemurafenib-resistant melanoma cells (A375/R cells) proliferation ability and its mechanism. Cell activity was assessed using the cell counting kit-8 (CCK-8) method. Flow cytometry assay was used to assess cell apoptosis and cell cycle. A xenograft model was used to analyze the inhibitory effects of evodiamine on tumor growth. Bioinformatics analyses, network pharmacology, and molecular docking were used to explore the potential mechanism of evodiamine in vemurafenib-resistant melanoma. RT-qPCR and Western blotting were performed to reveal the molecular mechanism. The alkaloid extract of the fruit of tetradium, evodiamine showed the strongest tumor inhibitory effect on vemurafenib-resistant melanoma cells compared to treatment with vemurafenib alone. Evodiamine inhibited vemurafenib-resistant melanoma cell growth, proliferation, and induced apoptosis, conforming to a dose-effect relationship and time-effect relationship. Results from network pharmacology and molecular docking suggested that evodiamine might interact with IRS4 to suppress growth of human vemurafenib-resistant melanoma cells. Interestingly, evodiamine suppressed IRS4 expression and then inhibited PI3K/AKT signaling pathway, and thus had the therapeutic action on vemurafenib-resistant melanoma.
Collapse
Affiliation(s)
- Xingxian Guo
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Shiying Huang
- Department of Clinical Laboratory, Zigong First People's Hospital, Zigong, China
| | - Yonghong Zhang
- Chongqing Engineering Research Center for Clinical Big-Data and Drug Evaluation Medical Data Science Academy, Chongqing Medical University, Chongqing, China
| | - Hong Wang
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Department of Pharmacy, Chongqing Health Center for Women and Children), Chongqing, China
| | - Lisha Li
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400010, China
| | - Jianhua Ran
- Neuroscience Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Dilong Chen
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Faculty of Basic Medical Sciences, Chongqing Three Gorges Medical College, Wanzhou, 404100, China
- NMPA Key Laboratory for Quality Monitoring of Narcotic Drugs and Psychotropic Substances, Chongqing Institute for Food and Drug Control, Chongqing, China
| | - Xiaopeng Li
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Faculty of Basic Medical Sciences, Chongqing Three Gorges Medical College, Wanzhou, 404100, China.
| | - Jing Li
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
17
|
Shah V, Panchal V, Shah A, Vyas B, Agrawal S, Bharadwaj S. Immune checkpoint inhibitors in metastatic melanoma therapy (Review). MEDICINE INTERNATIONAL 2024; 4:13. [PMID: 38410760 PMCID: PMC10895472 DOI: 10.3892/mi.2024.137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/26/2024] [Indexed: 02/28/2024]
Abstract
An increase in the incidence of melanoma has been observed in recent decades, which poses a significant challenge due to its poor prognosis in the advanced and metastatic stages. Previously, chemotherapy and high doses of interleukin-2 were available treatments for melanoma; however, they offered limited survival benefits and were associated with severe toxicities. The treatment of metastatic melanoma has been transformed by new developments in immunotherapy. Immune checkpoint inhibitors (ICIs), monoclonal antibodies that target cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), programmed cell death protein 1 (PD-1) and its ligand, PDL-1, have emerged as promising therapeutic options. Commonly used ICIs, such as ipilimumab, nivolumab and pembrolizumab, have been found to be associated with an improved median overall survival, recurrence-free survival and response rates compared to traditional chemotherapies. Combination therapies involving different types of ICIs, such as anti-PD1 with anti-CTLA-4, have further enhanced the overall survival and response rates by targeting various phases of T-cell activation. Additionally, the development of novel biomarkers has facilitated the assessment of responses to ICI therapy, with tissue and serum-based prognostic and predictive biomarkers now available. The increased response observed with ICIs also provides potential for immune-related adverse effects on various organ systems. Further research is required to evaluate the efficacy and safety of various combinations of ICIs, while ongoing clinical trials explore the potential of newer ICIs. Concerns regarding the development of resistance to ICIs also warrant attention. The present review summarizes and discusses the advent of ICIs with a marked significant breakthrough in the treatment of metastatic melanoma, providing improved outcomes compared to traditional therapies.
Collapse
Affiliation(s)
- Vedant Shah
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Viraj Panchal
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Abhi Shah
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Bhavya Vyas
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Siddharth Agrawal
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Sanket Bharadwaj
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| |
Collapse
|
18
|
Celeste FV, Powers S. Induction of Multiple Alternative Mitogenic Signaling Pathways Accompanies the Emergence of Drug-Tolerant Cancer Cells. Cancers (Basel) 2024; 16:1001. [PMID: 38473364 PMCID: PMC10930612 DOI: 10.3390/cancers16051001] [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: 01/30/2024] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Drug resistance can evolve from a subpopulation of cancer cells that initially survive drug treatment and then gradually form a pool of drug-tolerant cells. Several studies have pinpointed the activation of a specific bypass pathway that appears to provide the critical therapeutic target for preventing drug tolerance. Here, we take a systems-biology approach, using proteomics and genomics to examine the development of drug tolerance to EGFR inhibitors in EGFR-mutant lung adenocarcinoma cells and BRAF inhibitors in BRAF-mutant melanoma cells. We found that there are numerous alternative mitogenic pathways that become activated in both cases, including YAP, STAT3, IGFR1, and phospholipase C (PLC)/protein kinase C (PKC) pathways. Our results suggest that an effective therapeutic strategy to prevent drug tolerance will need to take multiple alternative mitogenic pathways into account rather than focusing on one specific pathway.
Collapse
Affiliation(s)
- Frank V. Celeste
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11794, USA
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Scott Powers
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11794, USA
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
19
|
Castillo-Ferrer C, Marguet T, Vanwonterghem L, Erbek S, Chuffart F, Mouret S, Messai ST, Gauchez AS, Coll JL, Charles J, Hurbin A, Martel-Frachet V. Serum IGF1 Is a Prognostic Marker for Resistance to Targeted Therapies and a Predictive Marker for Anti-IGF1 Receptor Therapy in Melanoma. J Invest Dermatol 2024; 144:422-426.e8. [PMID: 37580013 DOI: 10.1016/j.jid.2023.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 08/16/2023]
Affiliation(s)
- Camila Castillo-Ferrer
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble Alpes University, Grenoble, France; École Pratique des Hautes Études, PSL Research University, Paris, France
| | - Théo Marguet
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble Alpes University, Grenoble, France; Dermatology, Allergology & Photobiology Department, Grenoble Alpes University Hospital, Grenoble Alpes University, Grenoble, France
| | - Laetitia Vanwonterghem
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble Alpes University, Grenoble, France
| | - Sule Erbek
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble Alpes University, Grenoble, France; École Pratique des Hautes Études, PSL Research University, Paris, France
| | - Florent Chuffart
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble Alpes University, Grenoble, France
| | - Stéphane Mouret
- Dermatology, Allergology & Photobiology Department, Grenoble Alpes University Hospital, Grenoble Alpes University, Grenoble, France
| | - Sabiha Trabelsi Messai
- Dermatology, Allergology & Photobiology Department, Grenoble Alpes University Hospital, Grenoble Alpes University, Grenoble, France
| | - Anne-Sophie Gauchez
- Biology and Pathology Institute, Grenoble Alpes University Hospital, Grenoble Alpes University, Grenoble, France
| | - Jean-Luc Coll
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble Alpes University, Grenoble, France
| | - Julie Charles
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble Alpes University, Grenoble, France; Dermatology, Allergology & Photobiology Department, Grenoble Alpes University Hospital, Grenoble Alpes University, Grenoble, France
| | - Amandine Hurbin
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble Alpes University, Grenoble, France.
| | - Véronique Martel-Frachet
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble Alpes University, Grenoble, France; École Pratique des Hautes Études, PSL Research University, Paris, France
| |
Collapse
|
20
|
Jadav M, Solanki R, Patel S, Pooja D, Kulhari H. Development of thiolated xanthan gum-stearylamine conjugate based mucoadhesive system for the delivery of biochanin-A to melanoma cells. Int J Biol Macromol 2024; 257:128693. [PMID: 38092110 DOI: 10.1016/j.ijbiomac.2023.128693] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Recently, instead of creating new active compounds, scientists have been working to increase the bioavailability and residence time of existing drugs by modifying the characteristics of the delivery systems. In the present study, a novel mucoadhesive bioconjugate (SN-XG-SH) was synthesized by functionalizing a polysaccharide xanthan gum (XG) with cysteamine hydrochloride (CYS) and a lipid stearylamine (SN). FTIR, CHNS and 1H NMR studies confirmed the successful synthesis of SN-XG-SH. Mucoadhesion of the thiolated XG was enhanced and evaluated by different methods. Disulfide bond formation between thiolated XG and skin mucus enhances mucoadhesive behavior. The mucoadhesive bioconjugate was used to prepare nanoparticles for the delivery of hydrophobic biochanin-A (Bio-A) for the treatment of melanoma. The thiolated xanthan gum nanoparticles also demonstrated high drug entrapment efficiency, sustained drug release, and high storage stability. The drug loaded nanoparticles (Bio-A@TXNPs) significantly improved the cytotoxicity of Bio-A against human epidermoid cancer cells (A431 cells) by inducing apoptosis and changing mitochondrial membrane potential. In conclusion, thiolation of XG improves its mucoadhesive properties and prolongs the release of Bio-A. Thus, thiolated XG conjugate has a high potential for use as a bioadhesive agent in controlled and localised delivery of drugs in different skin diseases including melanoma.
Collapse
Affiliation(s)
- Mahima Jadav
- School of Nano Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India
| | - Raghu Solanki
- School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India
| | - Sunita Patel
- School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India
| | - Deep Pooja
- School of Pharmacy, National Forensic Science University, Gandhinagar, Gujarat 382007, India.
| | - Hitesh Kulhari
- School of Nano Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India.
| |
Collapse
|
21
|
Tzenaki N, Xenou L, Goulielmaki E, Tsapara A, Voudouri I, Antoniou A, Valianatos G, Tzardi M, De Bree E, Berdiaki A, Makrigiannakis A, Papakonstanti EA. A combined opposite targeting of p110δ PI3K and RhoA abrogates skin cancer. Commun Biol 2024; 7:26. [PMID: 38182748 PMCID: PMC10770346 DOI: 10.1038/s42003-023-05639-8] [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: 03/07/2023] [Accepted: 11/27/2023] [Indexed: 01/07/2024] Open
Abstract
Malignant melanoma is the most aggressive and deadly skin cancer with an increasing incidence worldwide whereas SCC is the second most common non-melanoma human skin cancer with limited treatment options. Here we show that the development and metastasis of melanoma and SCC cancers can be blocked by a combined opposite targeting of RhoA and p110δ PI3K. We found that a targeted induction of RhoA activity into tumours by deletion of p190RhoGAP-a potent inhibitor of RhoA GTPase-in tumour cells together with adoptive macrophages transfer from δD910A/D910A mice in mice bearing tumours with active RhoA abrogated growth progression of melanoma and SCC tumours. Τhe efficacy of this combined treatment is the same in tumours lacking activating mutations in BRAF and in tumours harbouring the most frequent BRAF(V600E) mutation. Furthermore, the efficiency of this combined treatment is associated with decreased ATX expression in tumour cells and tumour stroma bypassing a positive feedback expression of ATX induced by direct ATX pharmacological inactivation. Together, our findings highlight the importance of targeting cancer cells and macrophages for skin cancer therapy, emerge a reverse link between ATX and RhoA and illustrate the benefit of p110δ PI3K inhibition as a combinatorial regimen for the treatment of skin cancers.
Collapse
Affiliation(s)
- Niki Tzenaki
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Lydia Xenou
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Evangelia Goulielmaki
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Anna Tsapara
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Irene Voudouri
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Angelika Antoniou
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - George Valianatos
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Maria Tzardi
- Department of Pathology, School of Medicine, University of Crete, University Hospital, Heraklion, Greece
| | - Eelco De Bree
- Department of Surgical Oncology, School of Medicine, University of Crete, University Hospital, Heraklion, Greece
| | - Aikaterini Berdiaki
- Department of Obstetrics and Gynaecology, School of Medicine, University of Crete, University Hospital, Heraklion, Greece
| | - Antonios Makrigiannakis
- Department of Obstetrics and Gynaecology, School of Medicine, University of Crete, University Hospital, Heraklion, Greece
| | | |
Collapse
|
22
|
Eroglu Z, Chen YA, Smalley I, Li J, Markowitz JK, Brohl AS, Tetteh L, Taylor H, Sondak VK, Khushalani NI, Smalley KSM. Combined BRAF, MEK, and heat-shock protein 90 inhibition in advanced BRAF V600-mutant melanoma. Cancer 2024; 130:232-243. [PMID: 37776537 DOI: 10.1002/cncr.35029] [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: 06/21/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 10/02/2023]
Abstract
BACKGROUND Resistance to BRAF and MEK inhibitors in BRAF V600-mutant melanoma is common. Multiple resistance mechanisms involve heat-shock protein 90 (HSP90) clients, and a phase 1 study of vemurafenib with the HSP90 inhibitor XL888 in patients with advanced melanoma showed activity equivalent to that of BRAF and MEK inhibitors. METHODS Vemurafenib (960 mg orally twice daily) and cobimetinib (60 mg orally once daily for 21 of 28 days) with escalating dose cohorts of XL888 (30, 45, 60, or 90 mg orally twice weekly) was investigated in a phase 1 trial of advanced melanoma, with a modified Ji dose-escalation design. RESULTS Twenty-five patients were enrolled. After two dose-limiting toxicities (DLTs) (rash and acute kidney injury) in the first cohort, lower doses of vemurafenib (720 mg) and cobimetinib (40 mg) were investigated with the same XL888 doses. Three DLTs (rash) were observed in 12 patients in the XL888 60-mg cohort, and this was determined as the maximum tolerated dose. Objective responses were observed in 19 patients (76%), and the median progression-free survival was 7.6 months, with a 5-year progression-free survival rate of 20%. The median overall survival was 41.7 months, with a 5-year overall survival rate of 37%. Single-cell RNA sequencing was performed on baseline and on-treatment biopsies; treatment was associated with increased immune cell influx (CD4-positive and CD8-positive T cells) and decreased melanoma cells. CONCLUSIONS Combined vemurafenib and cobimetinib plus XL888 had significant toxicity, requiring frequent dose reductions, which may have contributed to the relatively low progression-free survival despite a high tumor response rate. Given overlapping toxicities, caution must be used when combining HSP90 inhibitors with BRAF and MEK inhibitors.
Collapse
Affiliation(s)
- Zeynep Eroglu
- Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Y Ann Chen
- Department of Biostatistics and Bioinformatics, The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Inna Smalley
- Department of Cancer Physiology, The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Jiannong Li
- Department of Biostatistics and Bioinformatics, The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Joseph K Markowitz
- Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Andrew S Brohl
- Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Leticia Tetteh
- Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Hayley Taylor
- Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Vernon K Sondak
- Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Nikhil I Khushalani
- Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Keiran S M Smalley
- Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
- Department of Tumor Biology, The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| |
Collapse
|
23
|
Wang P, Laster K, Jia X, Dong Z, Liu K. Targeting CRAF kinase in anti-cancer therapy: progress and opportunities. Mol Cancer 2023; 22:208. [PMID: 38111008 PMCID: PMC10726672 DOI: 10.1186/s12943-023-01903-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/16/2023] [Indexed: 12/20/2023] Open
Abstract
The RAS/mitogen-activated protein kinase (MAPK) signaling cascade is commonly dysregulated in human malignancies by processes driven by RAS or RAF oncogenes. Among the members of the RAF kinase family, CRAF plays an important role in the RAS-MAPK signaling pathway, as well as in the progression of cancer. Recent research has provided evidence implicating the role of CRAF in the physiological regulation and the resistance to BRAF inhibitors through MAPK-dependent and MAPK-independent mechanisms. Nevertheless, the effectiveness of solely targeting CRAF kinase activity remains controversial. Moreover, the kinase-independent function of CRAF may be essential for lung cancers with KRAS mutations. It is imperative to develop strategies to enhance efficacy and minimize toxicity in tumors driven by RAS or RAF oncogenes. The review investigates CRAF alterations observed in cancers and unravels the distinct roles of CRAF in cancers propelled by diverse oncogenes. This review also seeks to summarize CRAF-interacting proteins and delineate CRAF's regulation across various cancer hallmarks. Additionally, we discuss recent advances in pan-RAF inhibitors and their combination with other therapeutic approaches to improve treatment outcomes and minimize adverse effects in patients with RAF/RAS-mutant tumors. By providing a comprehensive understanding of the multifaceted role of CRAF in cancers and highlighting the latest developments in RAF inhibitor therapies, we endeavor to identify synergistic targets and elucidate resistance pathways, setting the stage for more robust and safer combination strategies for cancer treatment.
Collapse
Affiliation(s)
- Penglei Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, 450052, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Kyle Laster
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Xuechao Jia
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, 450052, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, 450052, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China.
- Department of Pathophysiology, School of Basic Medical Sciences, China-US (Henan) Hormel Cancer Institute, AMS, College of Medicine, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China.
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, 450052, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China.
- Department of Pathophysiology, School of Basic Medical Sciences, China-US (Henan) Hormel Cancer Institute, AMS, College of Medicine, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China.
- Basic Medicine Sciences Research Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
24
|
Bahar ME, Kim HJ, Kim DR. Targeting the RAS/RAF/MAPK pathway for cancer therapy: from mechanism to clinical studies. Signal Transduct Target Ther 2023; 8:455. [PMID: 38105263 PMCID: PMC10725898 DOI: 10.1038/s41392-023-01705-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/03/2023] [Accepted: 11/12/2023] [Indexed: 12/19/2023] Open
Abstract
Metastatic dissemination of solid tumors, a leading cause of cancer-related mortality, underscores the urgent need for enhanced insights into the molecular and cellular mechanisms underlying metastasis, chemoresistance, and the mechanistic backgrounds of individuals whose cancers are prone to migration. The most prevalent signaling cascade governed by multi-kinase inhibitors is the mitogen-activated protein kinase (MAPK) pathway, encompassing the RAS-RAF-MAPK kinase (MEK)-extracellular signal-related kinase (ERK) pathway. RAF kinase is a primary mediator of the MAPK pathway, responsible for the sequential activation of downstream targets, such as MEK and the transcription factor ERK, which control numerous cellular and physiological processes, including organism development, cell cycle control, cell proliferation and differentiation, cell survival, and death. Defects in this signaling cascade are associated with diseases such as cancer. RAF inhibitors (RAFi) combined with MEK blockers represent an FDA-approved therapeutic strategy for numerous RAF-mutant cancers, including melanoma, non-small cell lung carcinoma, and thyroid cancer. However, the development of therapy resistance by cancer cells remains an important barrier. Autophagy, an intracellular lysosome-dependent catabolic recycling process, plays a critical role in the development of RAFi resistance in cancer. Thus, targeting RAF and autophagy could be novel treatment strategies for RAF-mutant cancers. In this review, we delve deeper into the mechanistic insights surrounding RAF kinase signaling in tumorigenesis and RAFi-resistance. Furthermore, we explore and discuss the ongoing development of next-generation RAF inhibitors with enhanced therapeutic profiles. Additionally, this review sheds light on the functional interplay between RAF-targeted therapies and autophagy in cancer.
Collapse
Affiliation(s)
- Md Entaz Bahar
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Hyun Joon Kim
- Department of Anatomy and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea.
| |
Collapse
|
25
|
Chapdelaine AG, Ku GC, Sun G, Ayrapetov MK. The Targeted Degradation of BRAF V600E Reveals the Mechanisms of Resistance to BRAF-Targeted Treatments in Colorectal Cancer Cells. Cancers (Basel) 2023; 15:5805. [PMID: 38136350 PMCID: PMC10741866 DOI: 10.3390/cancers15245805] [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: 11/07/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
The BRAF V600E mutation is frequently found in cancer. It activates the MAPK pathway and promotes cancer cell proliferation, making BRAF an excellent target for anti-cancer therapy. While BRAF-targeted therapy is highly effective for melanoma, it is often ineffective against other cancers harboring the BRAF mutation. In this study, we evaluate the effectiveness of a proteolysis targeting chimera (PROTAC), SJF-0628, in directing the degradation of mutated BRAF across a diverse panel of cancer cells and determine how these cells respond to the degradation. SJF-0628 treatment results in the degradation of BRAF V600E and a decrease in Mek activation in all cell lines tested, but the effects of the treatment on cell signaling and cell proliferation are cell-line-specific. First, BRAF degradation killed DU-4475 and Colo-205 cells via apoptosis but only partially inhibited the proliferation of other cancer cell lines. Second, SJF-0628 treatment resulted in co-degradation of MEK in Colo-205 cells but did not have the same effect in other cell lines. Finally, cell lines partially inhibited by BRAF degradation also contain other oncogenic drivers, making them multi-driver cancer cells. These results demonstrate the utility of a PROTAC to direct BRAF degradation and reveal that multi-driver oncogenesis renders some colorectal cancer cells resistant to BRAF-targeted treatment.
Collapse
Affiliation(s)
| | | | - Gongqin Sun
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA; (A.G.C.); (G.C.K.)
| | | |
Collapse
|
26
|
Niessner H, Hüsch A, Kosnopfel C, Meinhardt M, Westphal D, Meier F, Schilling B, Sinnberg T. Exploring the In Vitro and In Vivo Therapeutic Potential of BRAF and MEK Inhibitor Combination in NRAS-Mutated Melanoma. Cancers (Basel) 2023; 15:5521. [PMID: 38067230 PMCID: PMC10705743 DOI: 10.3390/cancers15235521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 10/16/2024] Open
Abstract
INTRODUCTION Patients with NRAS-mutant metastatic melanoma often have an aggressive disease requiring a fast-acting, effective therapy. The MEK inhibitor binimetinib shows an overall response rate of 15% in patients with NRAS-mutant melanoma, providing a backbone for combination strategies. Our previous studies demonstrated that in NRAS-mutant melanoma, the antitumor activity of the MEK inhibitor binimetinib was significantly potentiated by the BRAFV600E/K inhibitor encorafenib through the induction of ER stress, leading to melanoma cell death by apoptotic mechanisms. Encorafenib combined with binimetinib was well tolerated in a phase III trial showing potent antitumor activity in BRAF-mutant melanoma, making a rapid evaluation in NRAS-mutant melanoma imminently feasible. These data provide a mechanistic rationale for the evaluation of binimetinib combined with encorafenib in preclinical and clinical studies on NRAS-mutant metastatic melanoma. METHODS The combination of BRAFi plus MEKi was tested in a monolayer culture of patient-derived cell lines and in corresponding patient-derived tissue slice cultures of NRAS-mutant melanoma. To investigate the treatment in vivo, NSG (NOD. Cg-PrkdcscidIl2rgtm1Wjl/SzJ) mice were subcutaneously injected with three different BRAF wild-type melanoma models harboring oncogenic NRAS mutations and treated orally with encorafenib (6 mg/kg body weight, daily) with or without binimetinib (8 mg/kg body weight, twice daily). In parallel, an individual healing attempt was carried out by treating one patient with an NRAS-mutated tumor. RESULTS Encorafenib was able to enhance the inhibitory effect on cell growth of binimetinib only in the cell line SKMel147 in vitro. It failed to enhance the apoptotic effect found in two other NRAS-mutated cell lines. Encorafenib led to a hyperactivation of ERK which could be reduced with the combinational treatment. In two of the three patient-derived tissue slice culture models of NRAS-mutant melanomas, a slight tendency of a combinatorial effect was seen which was not significant. Encorafenib showed a slight induction of the ER stress genes ATF4, CHOP, and NUPR1. The combinational treatment was able to enhance this effect, but not significantly. In the mouse model, the combination therapy of encorafenib with binimetinib resulted in reduced tumor growth compared to the control and encorafenib groups; however, the best effect in terms of tumor growth inhibition was measured in the binimetinib therapy group. The therapy showed no effect in an individual healing attempt for a patient suffering from metastatic, therapy-refractory NRAS-mutated melanoma. CONCLUSION In in vitro and ex vivo settings, the combination therapy was observed to elicit a response; however, it did not amplify the efficacy observed with binimetinib alone, whereas in a patient, the combinational treatment remained ineffective. The preclinical in vivo data showed no increased combinatorial effect. However, the in vivo effect of binimetinib as monotherapy was unexpectedly high in the tested regimen. Nevertheless, binimetinib proved to be advantageous in the treatment of melanoma in vivo and led to high rates of apoptosis in vitro; hence, it still seems to be a good base for combination with other substances in the treatment of patients with NRAS-mutant melanoma.
Collapse
Affiliation(s)
- Heike Niessner
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstr. 25, 72076 Tuebingen, Germany;
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, 72076 Tuebingen, Germany
| | - Anna Hüsch
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstr. 25, 72076 Tuebingen, Germany;
| | - Corinna Kosnopfel
- Department of Hematology, Oncology and Pneumology, University Hospital Muenster, 48149 Muenster, Germany;
| | - Matthias Meinhardt
- Department of Pathology, Medical Faculty and University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany;
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany; (D.W.); (F.M.)
| | - Dana Westphal
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany; (D.W.); (F.M.)
- Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, 01307 Dresden, Germany
| | - Friedegund Meier
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany; (D.W.); (F.M.)
- Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, 01307 Dresden, Germany
- Center for Regenerative Therapies Dresden, TU Dresden, 01307 Dresden, Germany
| | - Bastian Schilling
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany;
| | - Tobias Sinnberg
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstr. 25, 72076 Tuebingen, Germany;
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, 72076 Tuebingen, Germany
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
27
|
Harmange G, Hueros RAR, Schaff DL, Emert B, Saint-Antoine M, Kim LC, Niu Z, Nellore S, Fane ME, Alicea GM, Weeraratna AT, Simon MC, Singh A, Shaffer SM. Disrupting cellular memory to overcome drug resistance. Nat Commun 2023; 14:7130. [PMID: 37932277 PMCID: PMC10628298 DOI: 10.1038/s41467-023-41811-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/15/2023] [Indexed: 11/08/2023] Open
Abstract
Gene expression states persist for varying lengths of time at the single-cell level, a phenomenon known as gene expression memory. When cells switch states, losing memory of their prior state, this transition can occur in the absence of genetic changes. However, we lack robust methods to find regulators of memory or track state switching. Here, we develop a lineage tracing-based technique to quantify memory and identify cells that switch states. Applied to melanoma cells without therapy, we quantify long-lived fluctuations in gene expression that are predictive of later resistance to targeted therapy. We also identify the PI3K and TGF-β pathways as state switching modulators. We propose a pretreatment model, first applying a PI3K inhibitor to modulate gene expression states, then applying targeted therapy, which leads to less resistance than targeted therapy alone. Together, we present a method for finding modulators of gene expression memory and their associated cell fates.
Collapse
Affiliation(s)
- Guillaume Harmange
- Cellular and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Raúl A Reyes Hueros
- Department of Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dylan L Schaff
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin Emert
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Michael Saint-Antoine
- Department of Electrical and Computer Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Laura C Kim
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zijian Niu
- Department of Chemistry, College of the Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physics, College of the Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Shivani Nellore
- Department of Biology, College of the Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
- The Wharton School, University of Pennsylvania, Philadelphia, PA, USA
| | - Mitchell E Fane
- Cancer Signaling and Microenvironment Research Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Gretchen M Alicea
- Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Ashani T Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health, Baltimore, MD, USA
- Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Abhyudai Singh
- Department of Electrical and Computer Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Sydney M Shaffer
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Herlyn M, Villanueva J. Twenty years of research in melanoma therapy-From "nothing works" to cures: A personal account. Pigment Cell Melanoma Res 2023; 36:583-587. [PMID: 37726985 PMCID: PMC11187729 DOI: 10.1111/pcmr.13133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/21/2023]
Affiliation(s)
- Meenhard Herlyn
- Molecular & Cellular Oncogenesis Program, Ellen and Ronald Caplan Cancer Center, The Wistar Institute, Philadelphia, PA
| | - Jessie Villanueva
- Molecular & Cellular Oncogenesis Program, Ellen and Ronald Caplan Cancer Center, The Wistar Institute, Philadelphia, PA
| |
Collapse
|
29
|
Chen W, Park JI. Tumor Cell Resistance to the Inhibition of BRAF and MEK1/2. Int J Mol Sci 2023; 24:14837. [PMID: 37834284 PMCID: PMC10573597 DOI: 10.3390/ijms241914837] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
BRAF is one of the most frequently mutated oncogenes, with an overall frequency of about 50%. Targeting BRAF and its effector mitogen-activated protein kinase kinase 1/2 (MEK1/2) is now a key therapeutic strategy for BRAF-mutant tumors, and therapies based on dual BRAF/MEK inhibition showed significant efficacy in a broad spectrum of BRAF tumors. Nonetheless, BRAF/MEK inhibition therapy is not always effective for BRAF tumor suppression, and significant challenges remain to improve its clinical outcomes. First, certain BRAF tumors have an intrinsic ability to rapidly adapt to the presence of BRAF and MEK1/2 inhibitors by bypassing drug effects via rewired signaling, metabolic, and regulatory networks. Second, almost all tumors initially responsive to BRAF and MEK1/2 inhibitors eventually acquire therapy resistance via an additional genetic or epigenetic alteration(s). Overcoming these challenges requires identifying the molecular mechanism underlying tumor cell resistance to BRAF and MEK inhibitors and analyzing their specificity in different BRAF tumors. This review aims to update this information.
Collapse
Affiliation(s)
| | - Jong-In Park
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| |
Collapse
|
30
|
Feng T, Tang Z, Karges J, Shen J, Jin C, Chen Y, Pan Y, He Y, Ji L, Chao H. Exosome camouflaged coordination-assembled Iridium(III) photosensitizers for apoptosis-autophagy-ferroptosis induced combination therapy against melanoma. Biomaterials 2023; 301:122212. [PMID: 37385136 DOI: 10.1016/j.biomaterials.2023.122212] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 07/01/2023]
Abstract
Melanoma represents the most fatal form of skin cancer due to its resistance mechanisms and high capacity for the development of metastases. Among other medicinal techniques, photodynamic therapy is receiving increasing attention. Despite promising results, the application of photodynamic therapy is inherently limited due to interference from melanin, poor tissue penetration of photosensitizers, low loading into drug delivery systems, and a lack of tumor selectivity. To overcome these limitations, herein, the coordination-driven assembly of Ir(III) complex photosensitizers with Fe(III) ions into nanopolymers for combined photodynamic therapy and chemodynamic therapy is reported. While remaining stable under physiological conditions, the nanopolymers dissociated in the tumor microenvironment. Upon exposure to light, the Ir(III) complexes produced singlet oxygen and superoxide anion radicals, inducing cell death by apoptosis and autophagy. The Fe(III) ions were reduced to Fe(II) upon depletion of glutathione and reduction of the GPX4 levels, triggering cell death by ferroptosis. To provide tumor selectivity, the nanopolymers were further camouflaged with exosomes. The generated nanoparticles were found to eradicate a melanoma tumor as well as inhibit the formation of metastases inside a mouse model.
Collapse
Affiliation(s)
- Tao Feng
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Zixin Tang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| | - Jinchao Shen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Chengzhi Jin
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Yu Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Yihang Pan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, PR China.
| | - Yulong He
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, PR China.
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, PR China; MOE Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, 400201, PR China.
| |
Collapse
|
31
|
Liu Y, Ruan H, Lu F, Peng H, Luan W. miR-224-5p acts as a tumour suppressor and reverses the resistance to BRAF inhibitor in melanoma through directly targeting PAK4 to block the MAPK pathway. Pathol Res Pract 2023; 249:154772. [PMID: 37611431 DOI: 10.1016/j.prp.2023.154772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/08/2023] [Accepted: 08/16/2023] [Indexed: 08/25/2023]
Abstract
miR-224-5p has been shown to play both an oncogene and tumour suppressor role in many human tumours. However, the role and molecular mechanism of miR-224-5p in cutaneous melanoma remains unclear. miR-224-5p levels were downregulated in melanoma tissue, and low miR-224-5p expression was an independent risk factor for melanoma patients. miR-224-5p blocked proliferation, epithelial-to-mesenchymal transition (EMT), invasion, migration in BRAF wild-type melanoma cell, and overcome acquired BRAFi resistance in VMF-resistant melanoma cells. miR-224-5p exerted its role by directly repressing PAK4 to block the downstream CRAF/MEK/ERK pathways. We demonstrated that miR-224-5p inhibited melanoma growth and metastasis in vivo though xenograft tumor and pulmonary metastasis assay. Thus, miR-224-5p/PAK4-mediated CRAF/MEK/ERK pathways have therapeutic potential in melanoma treatment.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Plastic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Hongru Ruan
- Department of Plastic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Feng Lu
- Department of Plastic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Huiyong Peng
- Department of Laboratory Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang Medical School of Nanjing Medical University, Zhenjiang, Jiangsu, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Wenkang Luan
- Department of Plastic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
32
|
Żołek T, Mazurek A, Grudzinski IP. In Silico Studies of Novel Vemurafenib Derivatives as BRAF Kinase Inhibitors. Molecules 2023; 28:5273. [PMID: 37446932 DOI: 10.3390/molecules28135273] [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: 04/29/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
BRAF inhibitors have improved the treatment of advanced or metastatic melanoma in patients that harbor a BRAFT1799A mutation. Because of new insights into the role of aberrant glycosylation in drug resistance, we designed and studied three novel vemurafenib derivatives possessing pentose-associated aliphatic ligands-methyl-, ethyl-, and isopropyl-ketopentose moieties-as potent BRAFV600E kinase inhibitors. The geometries of these derivatives were optimized using the density functional theory method. Molecular dynamic simulations were performed to find interactions between the ligands and BRAFV600E kinase. Virtual screening was performed to assess the fate of derivatives and their systemic toxicity, genotoxicity, and carcinogenicity. The computational mapping of the studied ligand-BRAFV600E complexes indicated that the central pyrrole and pyridine rings of derivatives were located within the hydrophobic ATP-binding site of the BRAFV600E protein kinase, while the pentose ring and alkyl chains were mainly included in hydrogen bonding interactions. The isopropyl-ketopentose derivative was found to bind the BRAFV600E oncoprotein with more favorable energy interaction than vemurafenib. ADME-TOX in silico studies showed that the derivatives possessed some desirable pharmacokinetic and toxicologic properties. The present results open a new avenue to study the carbohydrate derivatives of vemurafenib as potent BRAFV600E kinase inhibitors to treat melanoma.
Collapse
Affiliation(s)
- Teresa Żołek
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02 097 Warsaw, Poland
| | - Adam Mazurek
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02 097 Warsaw, Poland
| | - Ireneusz P Grudzinski
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02 097 Warsaw, Poland
| |
Collapse
|
33
|
Kim S, Carvajal R, Kim M, Yang HW. Kinetics of RTK activation determine ERK reactivation and resistance to dual BRAF/MEK inhibition in melanoma. Cell Rep 2023; 42:112570. [PMID: 37252843 DOI: 10.1016/j.celrep.2023.112570] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/31/2023] [Accepted: 05/12/2023] [Indexed: 06/01/2023] Open
Abstract
The combination of BRAF and MEK inhibitors (BRAFi/MEKi) has shown promising response rates in treating BRAF-mutant melanoma by inhibiting ERK activation. However, treatment efficacy is limited by the emergence of drug-tolerant persister cells (persisters). Here, we show that the magnitude and duration of receptor tyrosine kinase (RTK) activation determine ERK reactivation and persister development. Our single-cell analysis reveals that only a small subset of melanoma cells exhibits effective RTK and ERK activation and develops persisters, despite uniform external stimuli. The kinetics of RTK activation directly influence ERK signaling dynamics and persister development. These initially rare persisters form major resistant clones through effective RTK-mediated ERK activation. Consequently, limiting RTK signaling suppresses ERK activation and cell proliferation in drug-resistant cells. Our findings provide non-genetic mechanistic insights into the role of heterogeneity in RTK activation kinetics in ERK reactivation and BRAFi/MEKi resistance, suggesting potential strategies for overcoming drug resistance in BRAF-mutant melanoma.
Collapse
Affiliation(s)
- Sungsoo Kim
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Richard Carvajal
- Department of Medicine, Columbia University, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Minah Kim
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Hee Won Yang
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
34
|
Merat R. The human antigen R as an actionable super-hub within the network of cancer cell persistency and plasticity. Transl Oncol 2023; 35:101722. [PMID: 37352624 DOI: 10.1016/j.tranon.2023.101722] [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: 04/07/2023] [Revised: 05/30/2023] [Accepted: 06/12/2023] [Indexed: 06/25/2023] Open
Abstract
In this perspective article, a clinically inspired phenotype-driven experimental approach is put forward to address the challenge of the adaptive response of solid cancers to small-molecule targeted therapies. A list of conditions is derived, including an experimental quantitative assessment of cell plasticity and an information theory-based detection of in vivo dependencies, for the discovery of post-transcriptional druggable mechanisms capable of preventing at multiple levels the emergence of plastic dedifferentiated slow-proliferating cells. The approach is illustrated by the author's own work in the example case of the adaptive response of BRAFV600-melanoma to BRAF inhibition. A bench-to-bedside and back to bench effort leads to a therapeutic strategy in which the inhibition of the baseline activity of the interferon-γ-activated inhibitor of translation (GAIT) complex, incriminated in the expression insufficiency of the RNA-binding protein HuR in a minority of cells, results in the suppression of the plastic, intermittently slow-proliferating cells involved in the adaptive response. A similar approach is recommended for the validation of other classes of mechanisms that we seek to modulate to overcome this complex challenge of modern cancer therapy.
Collapse
Affiliation(s)
- Rastine Merat
- Dermato-Oncology Unit, Division of Dermatology, Geneva University Hospitals, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland.
| |
Collapse
|
35
|
Ma W, Tian M, Hu L, Ruan X, Zhang W, Zheng X, Gao M. Early Combined SHP2 Targeting Reverses the Therapeutic Resistance of Vemurafenib in Thyroid Cancer. J Cancer 2023; 14:1592-1604. [PMID: 37325052 PMCID: PMC10266257 DOI: 10.7150/jca.83853] [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: 02/25/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023] Open
Abstract
The BRAFV600E mutation is the most common oncogenic mutation in thyroid cancer, suggesting an aggressive subtype of thyroid cancer and poor prognosis. Vemurafenib, a selective inhibitor of BRAFV600E, may provide therapeutic benefit in various cancers including thyroid cancer. However, the prevalence of drug resistance remains a challenge because of the feedback activation of the MAPK/ERK and PI3K/AKT pathways. In treating thyroid cancer cells with vemurafenib, we have detected reactivation of the MAPK/ERK signaling pathway as a result of the release of multiple receptor tyrosine kinases (RTKs) from the negative feedback of ERK phosphorylation. SHP2 is an important target protein downstream of the RTK signaling pathway. Decreasing it through SHP2 knockdown or the use of an inhibitor of SHP2 (SHP099) was found to significantly increase the early sensitivity and reverse the late resistance to vemurafenib in BRAFV600E mutant thyroid cancer cells. Overall, our findings suggest that blocking SHP2 reverses the reactivation of the MAPK/ERK signaling pathway caused by the activation of RTKs and improves the sensitivity of thyroid cancer to vemurafenib, which has potential implications for mechanism-based early combination strategies to treat thyroid cancer.
Collapse
Affiliation(s)
- Weike Ma
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Mengran Tian
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Linfei Hu
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xianhui Ruan
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Wei Zhang
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiangqian Zheng
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Ming Gao
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Tianjin Union Medical Center, No.190 Jieyuan Road, Hongqiao District, Tianjin 300121, China
| |
Collapse
|
36
|
Nelson BE, Reddy NK, Huse JT, Amini B, Nardo M, Gouda M, Weathers SP, Subbiah V. Histological transformation to gliosarcoma with combined BRAF/MEK inhibition in BRAF V600E mutated glioblastoma. NPJ Precis Oncol 2023; 7:47. [PMID: 37231247 DOI: 10.1038/s41698-023-00398-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/12/2023] [Indexed: 05/27/2023] Open
Abstract
The identification of BRAF V600 mutation in multiple cancers beyond melanoma and the development of combined BRAF and MEK targeting agents have altered the landscape of tissue-agnostic precision oncology therapies with an impact on survival outcomes. Despite initial efficacy, resistance emerges, and it is pertinent to identify putative resistance mechanisms. We report a case of recurrent glioblastoma (GBM) harboring BRAF V600E alteration who initially responded to combined BRAF + MEK inhibition and subsequently developed treatment resistance by histological transformation to gliosarcoma and acquisition of oncogenic KRAS G12D and an NF1 L1083R mutation. This documented case represents an initial evidence of a developing phenomenon in cancer research as it provides the first evidence of an emergent KRAS G12D/NF1 L1083R aberration with histological transformation occurring concurrently with primary BRAF V600E-altered glioblastoma as a previously unrecognized acquired mechanism of resistance in the setting of combined BRAF and MEK inhibition. This novel finding not only sheds new light on the RAS/MAPK pathway but also highlights the potential for morphological transformation to gliosarcoma, underscoring the critical need for further investigation in this area.
Collapse
Affiliation(s)
- Blessie Elizabeth Nelson
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neha K Reddy
- Department of Internal Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Jason T Huse
- Department of Pathology and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Behrang Amini
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mirella Nardo
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mohamed Gouda
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shiao-Pei Weathers
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- MD Anderson Cancer Network, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
37
|
Inhibiting SCD expression by IGF1R during lorlatinib therapy sensitizes melanoma to ferroptosis. Redox Biol 2023; 61:102653. [PMID: 36889082 PMCID: PMC10009726 DOI: 10.1016/j.redox.2023.102653] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 01/16/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023] Open
Abstract
Induction of ferroptosis is an emerging strategy to suppress melanoma progression. Strategies to enhance the sensitivity to ferroptosis induction would be a major advance in melanoma therapy. Here, we used a drug synergy screen that combined a ferroptosis inducer, RSL3, with 240 anti-tumor drugs from the FDA-approved drug library and identified lorlatinib to synergize with RSL3 in melanoma cells. We further demonstrated that lorlatinib sensitized melanoma to ferroptosis through inhibiting PI3K/AKT/mTOR signaling axis and its downstream SCD expression. Moreover, we found that lorlatinib's target IGF1R, but not ALK or ROS1, was the major mediator of lorlatinib-mediated sensitivity to ferroptosis through targeting PI3K/AKT/mTOR signaling axis. Finally, lorlatinib treatment sensitized melanoma to GPX4 inhibition in preclinical animal models, and melanoma patients with low GPX4 and IGF1R expression in their tumors survived for longer period. Altogether, lorlatinib sensitizes melanoma to ferroptosis by targeting IGF1R-mediated PI3K/AKT/mTOR signaling axis, suggesting that combination with lorlatinib could greatly expand the utility of GPX4 inhibition to melanoma patients with IGF1R-proficient expression.
Collapse
|
38
|
Ruffini F, Ceci C, Atzori MG, Caporali S, Levati L, Bonmassar L, Cappellini GCA, D'Atri S, Graziani G, Lacal PM. TARGETING OF PDGF-C/NRP-1 AUTOCRINE LOOP AS A NEW STRATEGY FOR COUNTERACTING THE INVASIVENESS OF MELANOMA RESISTANT TO BRAF INHIBITORS. Pharmacol Res 2023; 192:106782. [PMID: 37127213 DOI: 10.1016/j.phrs.2023.106782] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 04/19/2023] [Accepted: 04/27/2023] [Indexed: 05/03/2023]
Abstract
Melanoma resistance to BRAF inhibitors (BRAFi) is often accompanied by a switch from a proliferative to an invasive phenotype. Therefore, the identification of signaling molecules involved in the development of metastatic properties by resistant melanoma cells is of primary importance. We have previously demonstrated that activation of neuropilin-1 (NRP-1) by platelet-derived growth factor (PDGF)-C confers melanoma cells with an invasive behavior similar to that of BRAFi resistant tumors. Aims of the present study were to evaluate the role of PDGF-C/NRP-1 autocrine loop in the acquisition of an invasive and BRAFi-resistant phenotype by melanoma cells and the effect of its inhibition on drug resistance and extracellular matrix (ECM) invasion. Furthermore, we investigated whether PDGF-C serum levels were differentially modulated by drug treatment in metastatic melanoma patients responsive or refractory to BRAFi as a single agent or in combination with MEK inhibitors (MEKi). The results indicated that human melanoma cells resistant to BRAFi express higher levels of PDGF-C and NRP-1 as compared to their susceptible counterparts. Overexpression occurs early during development of drug resistance and contributes to the invasive properties of resistant cells. Accordingly, silencing of NRP-1 or PDGF-C reduces tumor cell invasiveness. Analysis of PDGF-C in the serum collected from patients treated with BRAFi or BRAFi+MEKi, showed that in responders PDGF-C levels decrease after treatment and raise again at tumor progression. Conversely, in non-responders treatment does not affect PDGF-C serum levels. Thus, blockade of NRP-1 activation by PDGF-C might represent a new therapeutic approach to counteract the invasiveness of BRAFi-resistant melanoma.
Collapse
Affiliation(s)
- Federica Ruffini
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Departmental Faculty of Medicine and Surgery, Saint Camillus International University of Health Sciences, Via di Sant'Alessandro, 8, 00131 Rome, Italy
| | | | | | | | | | | | | | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | |
Collapse
|
39
|
Talebi A, de Laat V, Spotbeen X, Dehairs J, Rambow F, Rogiers A, Vanderhoydonc F, Rizotto L, Planque M, Doglioni G, Motamedi S, Nittner D, Roskams T, Agostinis P, Bechter O, Boecxstaens V, Garmyn M, O'Farrell M, Wagman A, Kemble G, Leucci E, Fendt SM, Marine JC, Swinnen JV. Pharmacological induction of membrane lipid poly-unsaturation sensitizes melanoma to ROS inducers and overcomes acquired resistance to targeted therapy. J Exp Clin Cancer Res 2023; 42:92. [PMID: 37072838 PMCID: PMC10114329 DOI: 10.1186/s13046-023-02664-7] [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: 12/21/2022] [Accepted: 04/05/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND One of the key limitations of targeted cancer therapies is the rapid onset of therapy resistance. Taking BRAF-mutant melanoma as paradigm, we previously identified the lipogenic regulator SREBP-1 as a central mediator of resistance to MAPK-targeted therapy. Reasoning that lipogenesis-mediated alterations in membrane lipid poly-unsaturation lie at the basis of therapy resistance, we targeted fatty acid synthase (FASN) as key player in this pathway to evoke an exquisite vulnerability to clinical inducers of reactive oxygen species (ROS), thereby rationalizing a novel clinically actionable combination therapy to overcome therapy resistance. METHODS Using gene expression analysis and mass spectrometry-based lipidomics of BRAF-mutant melanoma cell lines, melanoma PDX and clinical data sets, we explored the association of FASN expression with membrane lipid poly-unsaturation and therapy-resistance. Next, we treated therapy-resistant models with a preclinical FASN inhibitor TVB-3664 and a panel of ROS inducers and performed ROS analysis, lipid peroxidation tests and real-time cell proliferation assays. Finally, we explored the combination of MAPK inhibitors, TVB-3664 and arsenic trioxide (ATO, as a clinically used ROS-inducer) in Mel006 BRAF mutant PDX as a gold model of therapy resistance and assessed the effect on tumor growth, survival and systemic toxicity. RESULTS We found that FASN expression is consistently increased upon the onset of therapy resistance in clinical melanoma samples, in cell lines and in Mel006 PDX and is associated with decreased lipid poly-unsaturation. Forcing lipid poly-unsaturation in therapy-resistant models by combining MAPK inhibition with FASN inhibition attenuated cell proliferation and rendered cells exquisitely sensitive to a host of ROS inducers. In particular, the triple combination of MAPK inhibition, FASN inhibition, and the clinical ROS-inducing compound ATO dramatically increased survival of Mel006 PDX models from 15 to 72% with no associated signs of toxicity. CONCLUSIONS We conclude that under MAPK inhibition the direct pharmacological inhibition of FASN evokes an exquisite vulnerability to inducers of ROS by increasing membrane lipid poly-unsaturation. The exploitation of this vulnerability by combining MAPK and/or FASN inhibitors with inducers of ROS greatly delays the onset of therapy resistance and increases survival. Our work identifies a clinically actionable combinatorial treatment for therapy-resistant cancer.
Collapse
Affiliation(s)
- Ali Talebi
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, LKI, KU Leuven, 3000, Leuven, Belgium
| | - Vincent de Laat
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, LKI, KU Leuven, 3000, Leuven, Belgium
| | - Xander Spotbeen
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, LKI, KU Leuven, 3000, Leuven, Belgium
| | - Jonas Dehairs
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, LKI, KU Leuven, 3000, Leuven, Belgium
| | - Florian Rambow
- Department of Applied Computational Cancer Research, Institute for AI in Medicine (IKIM), University Hospital Essen, Essen, Germany
- University of Duisburg-Essen, Essen, Germany
| | - Aljosja Rogiers
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, 3000, Leuven, Belgium
- Department of Oncology, Laboratory for Molecular Cancer Biology, KU Leuven, 3000, Leuven, Belgium
| | - Frank Vanderhoydonc
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, LKI, KU Leuven, 3000, Leuven, Belgium
| | - Lara Rizotto
- Department of Oncology, Laboratory for RNA Cancer Biology, LKI, KU Leuven, Leuven, Belgium
- Department of Oncology, Trace PDX Platform, LKI, KU Leuven, Leuven, Belgium
| | - Mélanie Planque
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, 3000, Leuven, Belgium
- Department of Oncology, Laboratory of Cellular Metabolism and Metabolic Regulation, LKI, KU Leuven, 3000, Leuven, Belgium
| | - Ginevra Doglioni
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, 3000, Leuven, Belgium
- Department of Oncology, Laboratory of Cellular Metabolism and Metabolic Regulation, LKI, KU Leuven, 3000, Leuven, Belgium
| | - Sahar Motamedi
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, LKI, KU Leuven, 3000, Leuven, Belgium
| | - David Nittner
- Histopathology Expertise Center, VIB-KU Leuven Center for Cancer Biology, 3000, Leuven, Belgium
| | - Tania Roskams
- Department of Imaging and Pathology, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Department of Cellular and Molecular Medicine, VIB-KU Leuven Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Oliver Bechter
- LKI, Department of General Medical Oncology, Department of Oncology, University Hospitals Leuven, KU, Leuven, Belgium
| | - Veerle Boecxstaens
- Department of Oncology, KU Leuven and Department of Surgical Oncology, UZ Leuven, Leuven, Belgium
| | - Marjan Garmyn
- Department of Oncology and Dermatology, Laboratory of Dermatology, University Hospitals Leuven, University of Leuven, Leuven, Belgium
| | - Marie O'Farrell
- Sagimet Biosciences, 155 Bovet Rd, San Mateo, CA, 94402, USA
| | - Alan Wagman
- 3-V Biosciences, Inc, 3715 Haven Ave, Menlo Park, CA, 94025, USA
| | - George Kemble
- Sagimet Biosciences, 155 Bovet Rd, San Mateo, CA, 94402, USA
| | - Eleonora Leucci
- Department of Oncology, Laboratory for RNA Cancer Biology, LKI, KU Leuven, Leuven, Belgium
- Department of Oncology, Trace PDX Platform, LKI, KU Leuven, Leuven, Belgium
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, 3000, Leuven, Belgium
- Department of Oncology, Laboratory of Cellular Metabolism and Metabolic Regulation, LKI, KU Leuven, 3000, Leuven, Belgium
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, 3000, Leuven, Belgium
- Department of Oncology, Laboratory for Molecular Cancer Biology, KU Leuven, 3000, Leuven, Belgium
| | - Johannes V Swinnen
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, LKI, KU Leuven, 3000, Leuven, Belgium.
| |
Collapse
|
40
|
Jin H, Wang L, Bernards R. Rational combinations of targeted cancer therapies: background, advances and challenges. Nat Rev Drug Discov 2023; 22:213-234. [PMID: 36509911 DOI: 10.1038/s41573-022-00615-z] [Citation(s) in RCA: 101] [Impact Index Per Article: 101.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 12/15/2022]
Abstract
Over the past two decades, elucidation of the genetic defects that underlie cancer has resulted in a plethora of novel targeted cancer drugs. Although these agents can initially be highly effective, resistance to single-agent therapies remains a major challenge. Combining drugs can help avoid resistance, but the number of possible drug combinations vastly exceeds what can be tested clinically, both financially and in terms of patient availability. Rational drug combinations based on a deep understanding of the underlying molecular mechanisms associated with therapy resistance are potentially powerful in the treatment of cancer. Here, we discuss the mechanisms of resistance to targeted therapies and how effective drug combinations can be identified to combat resistance. The challenges in clinically developing these combinations and future perspectives are considered.
Collapse
Affiliation(s)
- Haojie Jin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Liqin Wang
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - René Bernards
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
41
|
Alver TN, Heintz K, Hovig E, Bøe SL. Cooperative induction of receptor tyrosine kinases contributes to adaptive MAPK drug resistance in melanoma through the PI3K pathway. Cancer Rep (Hoboken) 2023; 6:e1736. [PMID: 36251678 PMCID: PMC9940011 DOI: 10.1002/cnr2.1736] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 08/05/2022] [Accepted: 09/07/2022] [Indexed: 02/22/2023] Open
Abstract
Vemurafenib-induced drug resistance in melanoma has been linked to receptor tyrosine kinase (RTK) upregulation. The MITF and SOX10 genes play roles as master regulators of melanocyte and melanoma development. Here, we aimed to explore the complex mechanisms behind the MITF/SOX10-controlled RTK-induced drug resistance in melanoma. To achieve this, we used a number of molecular techniques, including melanoma patient data from TCGA, vemurafenib-resistant melanoma cell lines, and knock-down studies. The melanoma cell lines were classified as proliferative or invasive based upon their MITF/AXL expression activity. We measured the change of expression activity for MITF/SOX10 and their receptor (AXL/ERBB3) and ligand (NRG1/GAS6) targets known to be involved in RTK-induced drug resistance after vemurafenib treatment. We find that melanoma cell lines characterized as proliferative (high MITF low AXL), transform into an invasive (low MITF, high AXL) cell state after vemurafenib resistance, indicating novel feedback loops and advanced compensatory regulation mechanisms between the master regulators, receptors, and ligands involved in vemurafenib-induced resistance. Together, our data disclose fine-tuned mechanisms involved in RTK-facilitated vemurafenib resistance that will be challenging to overcome by using single drug targeting strategies against melanoma.
Collapse
Affiliation(s)
- Tine Norman Alver
- Department of Tumor Biology, Institute for Cancer ResearchOslo University HospitalOsloNorway
- Department of Cancer Genetics, Institute for Cancer ResearchOslo University HospitalOsloNorway
- Faculty of MedicineUniversity of OsloOsloNorway
| | - Karen‐Marie Heintz
- Department of Tumor Biology, Institute for Cancer ResearchOslo University HospitalOsloNorway
- Faculty of MedicineUniversity of OsloOsloNorway
| | - Eivind Hovig
- Department of Tumor Biology, Institute for Cancer ResearchOslo University HospitalOsloNorway
- Department of InformaticsUniversity of OsloOsloNorway
| | - Sigurd L. Bøe
- Department of Medical BiochemistryOslo University Hospital, The Norwegian Radium HospitalOsloNorway
| |
Collapse
|
42
|
Rebecca VW, Xiao M, Kossenkov A, Godok T, Brown GS, Fingerman D, Alicea GM, Wei M, Ji H, Bravo J, Chen Y, Fane ME, Villanueva J, Nathanson K, Liu Q, Gopal YNV, Davies MA, Herlyn M. Dasatinib Resensitizes MAPK Inhibitor Efficacy in Standard-of-Care Relapsed Melanomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.20.524923. [PMID: 36711814 PMCID: PMC9882271 DOI: 10.1101/2023.01.20.524923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Resistance to combination BRAF/MEK inhibitor (BRAFi/MEKi) therapy arises in nearly every patient with BRAFV600E/K melanoma, despite promising initial responses. Achieving cures in this expanding BRAFi/MEKi-resistant cohort represents one of the greatest challenges to the field; few experience additional durable benefit from immunotherapy and no alternative therapies exist. To better personalize therapy in cancer patients to address therapy relapse, umbrella trials have been initiated whereby genomic sequencing of a panel of potentially actionable targets guide therapy selection for patients; however, the superior efficacy of such approaches remains to be seen. We here test the robustness of the umbrella trial rationale by analyzing relationships between genomic status of a gene and the downstream consequences at the protein level of related pathway, which find poor relationships between mutations, copy number amplification, and protein level. To profile candidate therapeutic strategies that may offer clinical benefit in the context of acquired BRAFi/MEKi resistance, we established a repository of patient-derived xenograft models from heavily pretreated patients with resistance to BRAFi/MEKi and/or immunotherapy (R-PDX). With these R-PDXs, we executed in vivo compound repurposing screens using 11 FDA-approved agents from an NCI-portfolio with pan-RTK, non-RTK and/or PI3K-mTOR specificity. We identify dasatinib as capable of restoring BRAFi/MEKi antitumor efficacy in ~70% of R-PDX tested. A systems-biology analysis indicates elevated baseline protein expression of canonical drivers of therapy resistance (e.g., AXL, YAP, HSP70, phospho-AKT) as predictive of MAPKi/dasatinib sensitivity. We therefore propose that dasatinib-based MAPKi therapy may restore antitumor efficacy in patients that have relapsed to standard-of-care therapy by broadly targeting proteins critical in melanoma therapy escape. Further, we submit that this experimental PDX paradigm could potentially improve preclinical evaluation of therapeutic modalities and augment our ability to identify biomarker-defined patient subsets that may respond to a given clinical trial.
Collapse
Affiliation(s)
- Vito W Rebecca
- The Wistar Institute, Philadelphia, PA, USA
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Min Xiao
- The Wistar Institute, Philadelphia, PA, USA
| | | | | | | | | | - Gretchen M Alicea
- The Wistar Institute, Philadelphia, PA, USA
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Meihan Wei
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Hongkai Ji
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jeremy Bravo
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Mitchell E Fane
- The Wistar Institute, Philadelphia, PA, USA
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | | | | | - Qin Liu
- The Wistar Institute, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
43
|
Malone CF, Kim M, Alexe G, Engel K, Forman AB, Robichaud A, Conway AS, Goodale A, Meyer A, Khalid D, Thayakumar A, Hatcher JM, Gray NS, Piccioni F, Stegmaier K. Transcriptional Antagonism by CDK8 Inhibition Improves Therapeutic Efficacy of MEK Inhibitors. Cancer Res 2023; 83:285-300. [PMID: 36398965 PMCID: PMC9938728 DOI: 10.1158/0008-5472.can-21-4309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 09/21/2022] [Accepted: 11/15/2022] [Indexed: 11/20/2022]
Abstract
Aberrant RAS/MAPK signaling is a common driver of oncogenesis that can be therapeutically targeted with clinically approved MEK inhibitors. Disease progression on single-agent MEK inhibitors is common, however, and combination therapies are typically required to achieve significant clinical benefit in advanced cancers. Here we focused on identifying MEK inhibitor-based combination therapies in neuroblastoma with mutations that activate the RAS/MAPK signaling pathway, which are rare at diagnosis but frequent in relapsed neuroblastoma. A genome-scale CRISPR-Cas9 functional genomic screen was deployed to identify genes that when knocked out sensitize RAS-mutant neuroblastoma to MEK inhibition. Loss of either CCNC or CDK8, two members of the mediator kinase module, sensitized neuroblastoma to MEK inhibition. Furthermore, small-molecule kinase inhibitors of CDK8 improved response to MEK inhibitors in vitro and in vivo in RAS-mutant neuroblastoma and other adult solid tumors. Transcriptional profiling revealed that loss of CDK8 or CCNC antagonized the transcriptional signature induced by MEK inhibition. When combined, loss of CDK8 or CCNC prevented the compensatory upregulation of progrowth gene expression induced by MEK inhibition. These findings propose a new therapeutic combination for RAS-mutant neuroblastoma and may have clinical relevance for other RAS-driven malignancies. SIGNIFICANCE Transcriptional adaptation to MEK inhibition is mediated by CDK8 and can be blocked by the addition of CDK8 inhibitors to improve response to MEK inhibitors in RAS-mutant neuroblastoma, a clinically challenging disease.
Collapse
Affiliation(s)
- Clare F. Malone
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Minjee Kim
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Gabriela Alexe
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Kathleen Engel
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alexandra B. Forman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Amanda Robichaud
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amy Saur Conway
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amy Goodale
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ashleigh Meyer
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Delan Khalid
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Allen Thayakumar
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - John M. Hatcher
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA,Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Nathanael S. Gray
- Department of Chemical and Systems Biology, ChEM-H, and Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | | | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA,Harvard Medical School, Boston, MA, USA,Corresponding author. Mailing address: Dana-Farber Cancer Institute, 360 Longwood Ave, LC6102, Boston, MA, 02215. Phone: (617) 632-4438
| |
Collapse
|
44
|
Hebron KE, Wan X, Roth JS, Liewehr DJ, Sealover NE, Frye WJ, Kim A, Stauffer S, Perkins OL, Sun W, Isanogle KA, Robinson CM, James A, Awasthi P, Shankarappa P, Luo X, Lei H, Butcher D, Smith R, Edmondson EF, Chen JQ, Kedei N, Peer CJ, Shern JF, Figg WD, Chen L, Hall MD, Difilippantonio S, Barr FG, Kortum RL, Robey RW, Vaseva AV, Khan J, Yohe ME. The Combination of Trametinib and Ganitumab is Effective in RAS-Mutated PAX-Fusion Negative Rhabdomyosarcoma Models. Clin Cancer Res 2023; 29:472-487. [PMID: 36322002 PMCID: PMC9852065 DOI: 10.1158/1078-0432.ccr-22-1646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/22/2022] [Accepted: 10/31/2022] [Indexed: 11/05/2022]
Abstract
PURPOSE PAX-fusion negative rhabdomyosarcoma (FN RMS) is driven by alterations in the RAS/MAP kinase pathway and is partially responsive to MEK inhibition. Overexpression of IGF1R and its ligands is also observed in FN RMS. Preclinical and clinical studies have suggested that IGF1R is itself an important target in FN RMS. Our previous studies revealed preclinical efficacy of the MEK1/2 inhibitor, trametinib, and an IGF1R inhibitor, BMS-754807, but this combination was not pursued clinically due to intolerability in preclinical murine models. Here, we sought to identify a combination of an MEK1/2 inhibitor and IGF1R inhibitor, which would be tolerated in murine models and effective in both cell line and patient-derived xenograft models of RAS-mutant FN RMS. EXPERIMENTAL DESIGN Using proliferation and apoptosis assays, we studied the factorial effects of trametinib and ganitumab (AMG 479), a mAb with specificity for human and murine IGF1R, in a panel of RAS-mutant FN RMS cell lines. The molecular mechanism of the observed synergy was determined using conventional and capillary immunoassays. The efficacy and tolerability of trametinib/ganitumab was assessed using a panel of RAS-mutated cell-line and patient-derived RMS xenograft models. RESULTS Treatment with trametinib and ganitumab resulted in synergistic cellular growth inhibition in all cell lines tested and inhibition of tumor growth in four of six models of RAS-mutant RMS. The combination had little effect on body weight and did not produce thrombocytopenia, neutropenia, or hyperinsulinemia in tumor-bearing SCID beige mice. Mechanistically, ganitumab treatment prevented the phosphorylation of AKT induced by MEK inhibition alone. Therapeutic response to the combination was observed in models without a mutation in the PI3K/PTEN axis. CONCLUSIONS We demonstrate that combined trametinib and ganitumab is effective in a genomically diverse panel of RAS-mutated FN RMS preclinical models. Our data also show that the trametinib/ganitumab combination likely has a favorable tolerability profile. These data support testing this combination in a phase I/II clinical trial for pediatric patients with relapsed or refractory RAS-mutated FN RMS.
Collapse
Affiliation(s)
- Katie E. Hebron
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892,Laboratory of Cell and Developmental Signaling, Center for Cancer Research, 8560 Progress Drive, Frederick, MD 21701
| | - Xiaolin Wan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Jacob S. Roth
- Early Translation Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD 20850
| | - David J. Liewehr
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Nancy E. Sealover
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Services, Bethesda, MD 20814
| | - William J.E. Frye
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892
| | - Angela Kim
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, 8560 Progress Drive, Frederick, MD 21701
| | - Stacey Stauffer
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, 8560 Progress Drive, Frederick, MD 21701
| | - Olivia L. Perkins
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Wenyue Sun
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892
| | - Kristine A. Isanogle
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Christina M. Robinson
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Amy James
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Parirokh Awasthi
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Priya Shankarappa
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Xiaoling Luo
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Haiyan Lei
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Donna Butcher
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Roberta Smith
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Elijah F. Edmondson
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Jin-Qiu Chen
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Noemi Kedei
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Cody J. Peer
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Jack F. Shern
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - W. Douglas Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Lu Chen
- Early Translation Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD 20850
| | - Matthew D. Hall
- Early Translation Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD 20850
| | - Simone Difilippantonio
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Frederic G. Barr
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892
| | - Robert L. Kortum
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Services, Bethesda, MD 20814
| | - Robert W. Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892
| | - Angelina V. Vaseva
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, Texas, USA
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892,Co-corresponding authors Correspondence: Marielle Yohe, M.D., Ph.D., Center for Cancer Research, National Cancer Institute, 8560 Progress Drive Room D3026, Frederick, MD 27101, Phone: (240) 760-7436,
| | - Marielle E. Yohe
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892,Laboratory of Cell and Developmental Signaling, Center for Cancer Research, 8560 Progress Drive, Frederick, MD 21701,Co-corresponding authors Correspondence: Marielle Yohe, M.D., Ph.D., Center for Cancer Research, National Cancer Institute, 8560 Progress Drive Room D3026, Frederick, MD 27101, Phone: (240) 760-7436,
| |
Collapse
|
45
|
Song Y, Bi Z, Liu Y, Qin F, Wei Y, Wei X. Targeting RAS-RAF-MEK-ERK signaling pathway in human cancer: Current status in clinical trials. Genes Dis 2023; 10:76-88. [PMID: 37013062 PMCID: PMC10066287 DOI: 10.1016/j.gendis.2022.05.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 04/23/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022] Open
Abstract
Molecular target inhibitors have been regularly approved by Food and Drug Administration (FDA) for tumor treatment, and most of them intervene in tumor cell proliferation and metabolism. The RAS-RAF-MEK-ERK pathway is a conserved signaling pathway that plays vital roles in cell proliferation, survival, and differentiation. The aberrant activation of the RAS-RAF-MEK-ERK signaling pathway induces tumors. About 33% of tumors harbor RAS mutations, while 8% of tumors are driven by RAF mutations. Great efforts have been dedicated to targeting the signaling pathway for cancer treatment in the past decades. In this review, we summarized the development of inhibitors targeting the RAS-RAF-MEK-ERK pathway with an emphasis on those used in clinical treatment. Moreover, we discussed the potential combinations of inhibitors that target the RAS-RAF-MEK-ERK signaling pathway and other signaling pathways. The inhibitors targeting the RAS-RAF-MEK-ERK pathway have essentially modified the therapeutic strategy against various cancers and deserve more attention in the current cancer research and treatment.
Collapse
Affiliation(s)
| | | | - Yu Liu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Furong Qin
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
46
|
Lehmann R, Rayner BS, Ziegler DS. Resistance mechanisms in BRAF V600E paediatric high-grade glioma and current therapeutic approaches. Front Oncol 2022; 12:1031378. [PMID: 36582791 PMCID: PMC9792688 DOI: 10.3389/fonc.2022.1031378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/25/2022] [Indexed: 12/15/2022] Open
Abstract
Paediatric high-grade gliomas (pHGG) are aggressive central nervous system tumours with a poor prognosis. BRAFV600E mutant pHGGs can be treated with targeted BRAF inhibitors, which have shown both preclinical activity and potent clinical efficacy. Unfortunately, the development of drug resistance results in disease relapse or progression and is the primary cause of treatment failure. While there is a lot of data to explain mechanisms of resistance in other BRAFV600E tumours, comparatively little is known about the mechanisms of BRAF inhibitor resistance in BRAFV600E pHGG. Recent literature has identified aberrations in members of the RAS/RAF/ERK pathway, the PI3K/AKT/MTOR pathway and the cell cycle as major contributors to the resistance profile. A range of novel therapies have been suggested to overcome BRAF inhibitor drug resistance in BRAFV600E pHGG. This review will discuss the current literature available for BRAF inhibitor resistant BRAFV600E pHGGs and provide an overview of the currently available and proposed therapies.
Collapse
Affiliation(s)
- R Lehmann
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
- School of Clinical Medicine, University of New South Wales (UNSW) Medicine & Health, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
| | - B S Rayner
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
- School of Clinical Medicine, University of New South Wales (UNSW) Medicine & Health, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
| | - D S Ziegler
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
- School of Clinical Medicine, University of New South Wales (UNSW) Medicine & Health, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| |
Collapse
|
47
|
Wu J, Lin Z. Non-Small Cell Lung Cancer Targeted Therapy: Drugs and Mechanisms of Drug Resistance. Int J Mol Sci 2022; 23:ijms232315056. [PMID: 36499382 PMCID: PMC9738331 DOI: 10.3390/ijms232315056] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
The advent of precision medicine has brought light to the treatment of non-small cell lung cancer (NSCLC), expanding the options for patients with advanced NSCLC by targeting therapy through genetic and epigenetic cues. Tumor driver genes in NSCLC patients have been uncovered one by one, including epidermal growth factor receptor (EGFR), mesenchymal lymphoma kinase (ALK), and receptor tyrosine kinase ROS proto-oncogene 1 (ROS1) mutants. Antibodies and inhibitors that target the critical gene-mediated signaling pathways that regulate tumor growth and development are anticipated to increase patient survival and quality of life. Targeted drugs continue to emerge, with as many as two dozen approved by the FDA, and chemotherapy and targeted therapy have significantly improved patient prognosis. However, resistance due to cancer drivers' genetic alterations has given rise to significant challenges in treating patients with metastatic NSCLC. Here, we summarized the main targeted therapeutic sites of NSCLC drugs and discussed their resistance mechanisms, aiming to provide new ideas for follow-up research and clues for the improvement of targeted drugs.
Collapse
|
48
|
Rubanov A, Berico P, Hernando E. Epigenetic Mechanisms Underlying Melanoma Resistance to Immune and Targeted Therapies. Cancers (Basel) 2022; 14:cancers14235858. [PMID: 36497341 PMCID: PMC9738385 DOI: 10.3390/cancers14235858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022] Open
Abstract
Melanoma is an aggressive skin cancer reliant on early detection for high likelihood of successful treatment. Solar UV exposure transforms melanocytes into highly mutated tumor cells that metastasize to the liver, lungs, and brain. Even upon resection of the primary tumor, almost thirty percent of patients succumb to melanoma within twenty years. Identification of key melanoma genetic drivers led to the development of pharmacological BRAFV600E and MEK inhibitors, significantly improving metastatic patient outcomes over traditional cytotoxic chemotherapy or pioneering IFN-α and IL-2 immune therapies. Checkpoint blockade inhibitors releasing the immunosuppressive effects of CTLA-4 or PD-1 proved to be even more effective and are the standard first-line treatment. Despite these major improvements, durable responses to immunotherapy and targeted therapy have been hindered by intrinsic or acquired resistance. In addition to gained or selected genetic alterations, cellular plasticity conferred by epigenetic reprogramming is emerging as a driver of therapy resistance. Epigenetic regulation of chromatin accessibility drives gene expression and establishes distinct transcriptional cell states. Here we review how aberrant chromatin, transcriptional, and epigenetic regulation contribute to therapy resistance and discuss how targeting these programs sensitizes melanoma cells to immune and targeted therapies.
Collapse
Affiliation(s)
- Andrey Rubanov
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Pietro Berico
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Eva Hernando
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
- Correspondence:
| |
Collapse
|
49
|
A Necroptosis-Related Gene Signature to Predict the Prognosis of Skin Cutaneous Melanoma. DISEASE MARKERS 2022; 2022:8232024. [DOI: 10.1155/2022/8232024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/26/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022]
Abstract
The prognosis of skin cutaneous melanoma (SKCM) remains poor, and patients with SKCM show a poor response to immunotherapy. Thus, we aimed to identify necroptosis-related biomarkers, which can help predict the prognosis of SKCM and improve the effectiveness of precision medicine. Data of SKCM were obtained from The Cancer Genome Atlas (TCGA) and GEO databases. TCGA samples were classified into two clusters by consensus clustering of necroptosis-related genes. Univariate Cox and least absolute shrinkage and selection operator regression analyses led to the identification of 11 genes, which were used to construct a prognostic model. GSE65904 was used as the test set. Principal component, t-distributed stochastic neighbor embedding, and Kaplan–Meier survival analyses indicated that samples in the train and test sets could be divided into two groups, with the high-risk group showing a worse prognosis. Univariate and multivariate Cox regression analyses were performed, and a nomogram, calibration curve, and time-dependent receiver operating characteristic curve were constructed to verify the efficacy of our model. The 1-, 3-, and 5-year areas under the receiver operating characteristic curves for the train set were 0.702, 0.663, and 0.701 and for the test set were 0.613, 0.627, and 0.637, respectively. Moreover, we performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses between the high- and low-risk groups. Single sample gene set enrichment analysis, immune cell infiltration analysis, tumor microenvironment scores, immune checkpoint analysis, and half-maximal inhibitory concentration prediction indicated that the high-risk group showed weaker antitumor immunity; further, the response to immune checkpoint inhibitors was worse, and the high-risk group was sensitive to fewer antitumor drugs. Tumor mutational burden analysis, Kaplan–Meier survival analysis, and correlation analysis between risk score and RNA stemness score revealed that the high-risk group with low tumor mutational burden and high RNA stemness score was potentially associated with poor prognosis. To conclude, our model, which was based on 11 necroptosis-related genes, could predict the prognosis of SKCM; in addition, it has guiding significance for the selection of clinical treatment and provides new research directions to enhance necroptosis against SKCM.
Collapse
|
50
|
Donati G, Amati B. MYC and therapy resistance in cancer: risks and opportunities. Mol Oncol 2022; 16:3828-3854. [PMID: 36214609 PMCID: PMC9627787 DOI: 10.1002/1878-0261.13319] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/08/2022] [Accepted: 10/06/2022] [Indexed: 12/24/2022] Open
Abstract
The MYC transcription factor, encoded by the c-MYC proto-oncogene, is activated by growth-promoting signals, and is a key regulator of biosynthetic and metabolic pathways driving cell growth and proliferation. These same processes are deregulated in MYC-driven tumors, where they become critical for cancer cell proliferation and survival. As other oncogenic insults, overexpressed MYC induces a series of cellular stresses (metabolic, oxidative, replicative, etc.) collectively known as oncogenic stress, which impact not only on tumor progression, but also on the response to therapy, with profound, multifaceted consequences on clinical outcome. On one hand, recent evidence uncovered a widespread role for MYC in therapy resistance in multiple cancer types, with either standard chemotherapeutic or targeted regimens. Reciprocally, oncogenic MYC imparts a series of molecular and metabolic dependencies to cells, thus giving rise to cancer-specific vulnerabilities that may be exploited to obtain synthetic-lethal interactions with novel anticancer drugs. Here we will review the current knowledge on the links between MYC and therapeutic responses, and will discuss possible strategies to overcome resistance through new, targeted interventions.
Collapse
Affiliation(s)
- Giulio Donati
- European Institute of Oncology (IEO) – IRCCSMilanItaly
| | - Bruno Amati
- European Institute of Oncology (IEO) – IRCCSMilanItaly
| |
Collapse
|