1
|
Ettienne EB, Grant-Kels JM, Striano P, Russo E, Neubauer D, Rose K. Melanoma and pediatric drug development: clinical progress vs. regulatory activism in minors - a narrative review. Expert Opin Pharmacother 2025; 26:595-603. [PMID: 40035212 DOI: 10.1080/14656566.2025.2475184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
INTRODUCTION Melanoma is the malignancy where, in recent years, drug treatment has massively improved prognosis and quality of life. Based on genetic analysis, we differentiate today melanomas caused by cumulative solar damage (CSD) from others. AREAS COVERED Conventional CSD-caused melanomas affect predominantly adults and occasionally adolescents. Spitz melanoma and melanoma arising in congenital nevi, the two other pediatric melanoma types, are not CSD-caused, are genetically different, have different prognoses, and need different treatment. In contrast to the improved situation in adult melanoma, regulatory demand for pediatric labels in minors has resulted in pointless and harmful studies and has obfuscated diagnosis and treatment. Modern communication facilitates a worldwide analysis of extremely rare diseases such as melanoma in minors. Regulatory demand for on-label treatment only is demonstrably wrong for pediatric melanomas. They are too rare for randomized controlled trials but nevertheless deserve effective treatment. EXPERT OPINION Adolescents with conventional, CSD-caused melanoma should be treated as adults. Their bodies are already mature. For other childhood melanomas, registries and consultations with clinical specialists are better options than dogmatically demanded regulatory studies.
Collapse
Affiliation(s)
| | - Jane M Grant-Kels
- Department of Dermatology, University of Connecticut Health Center, Farmington, CT, USA
| | | | - Emilio Russo
- Pharmacology, University of Magna Graecia, Catanzaro, Italy
| | - David Neubauer
- Department of Child, Adolescent & Developmental Neurology, University Childrens' Hospital, Ljubljana, Slovenia
| | - Klaus Rose
- klausrose Consulting, Riehen, Switzerland
| |
Collapse
|
2
|
Cui Z, Chen X, Zhai S, Wang Y, Hu C, Yuan B. Bibliometric and visual analysis of drug-specific immunotherapy from 1990 to 2024. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04073-3. [PMID: 40131388 DOI: 10.1007/s00210-025-04073-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025]
Abstract
Specific immunotherapy (SIT) is key in allergic diseases, tumor immunity, and autoimmune regulation. In recent years, the mechanism of action of drugs in SIT has attracted much attention, including the induction of hypersensitivity responses and modulation of immune tolerance. However, scientific challenges remain regarding their mechanism of action and optimization strategies. Studies on pharmacological SIT have been accumulated in the past, and there is an urgent need for bibliometric analyses to review and prospect these results for future academic development. Strict search criteria were developed to screen and download literature information from the Web of Science Core Collection. Six elements of the included literature were analyzed and visualized using Citespace, VOSviewer software, and the Bibliometrix package. A total of 682 publications related to the drug SIT were included in this study. The growth trend in the number of publications is evident and entering a new phase with great potential for the future, with the highest total number of citations in 1998. The countries and institutions with the most publications were the USA and the University of Genoa. The author who contributed the most to the field was Incorvaia, Cristoforo. Moreover, Bousquet J was the most influential author. Allergy was considered the leading core source journal, and the Journal of Allergy and Clinical Immunology is the most influential. The reference with the highest outbreak intensity is Roberts G, 2018, Allergy, V73, P765, https://doi.org/10.1111/all.13317 . The analysis of the keywords by the various metrics shows that the research hotspot is the tumor-associated SIT, the cutting-edge topic is the mechanism of action of the drug AIT, and the respiratory SIT application is the cutting-edge hot issue. Drug SIT has made good progress with the joint participation of global research institutions, scientists, and various journals. However, cooperation and communication are weak, and it is necessary to build a new cooperation mode to promote the globalization of research results. We have found the research hotspots and cutting-edge issues in this field, which will guide us to make breakthroughs in a more precise direction and play the important role of drug safety in safeguarding human life and health.
Collapse
Affiliation(s)
- Zhengjiu Cui
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaorui Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Siming Zhai
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuanyuan Wang
- Suqian Affiliated Hospital of Nanjing University of Chinese Medicine, Suqian, China
| | - Chanchan Hu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Bin Yuan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
3
|
Zielińska MK, Ciążyńska M, Sulejczak D, Rutkowski P, Czarnecka AM. Mechanisms of Resistance to Anti-PD-1 Immunotherapy in Melanoma and Strategies to Overcome It. Biomolecules 2025; 15:269. [PMID: 40001572 PMCID: PMC11853485 DOI: 10.3390/biom15020269] [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: 10/14/2024] [Revised: 12/22/2024] [Accepted: 01/10/2025] [Indexed: 02/27/2025] Open
Abstract
Resistance to anti-PD-1 therapy in melanoma remains a major obstacle in achieving effective and durable treatment outcomes, highlighting the need to understand and address the underlying mechanisms. The first key factor is innate anti-PD-1 resistance signature (IPRES), an expression of a group of genes associated with tumor plasticity and immune evasion. IPRES promotes epithelial-to-mesenchymal transition (EMT), increasing melanoma cells' invasiveness and survival. Overexpressed AXL, TWIST2, and WNT5a induce phenotypic changes. The upregulation of pro-inflammatory cytokines frequently coincides with EMT-related changes, further promoting a resistant and aggressive tumor phenotype. Inflamed tumor microenvironment may also drive the expression of resistance. The complexity of immune resistance development suggests that combination therapies are necessary to overcome it. Furthermore, targeting epigenetic regulation and exploring novel approaches such as miR-146a modulation may provide new strategies to counter resistance in melanoma.
Collapse
Affiliation(s)
- Magdalena K. Zielińska
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.K.Z.); (P.R.)
- Faculty of Medicine, Warsaw Medical University, 02-091 Warsaw, Poland
| | - Magdalena Ciążyńska
- Chemotherapy Unit and Day Chemotherapy Ward, Specialised Oncology Hospital, 97-200 Tomaszów Mazowiecki, Poland;
- Department of Dermatology, Paediatric Dermatology and Oncology Clinic, Medical University of Lodz, 91-347 Łódź, Poland
| | - Dorota Sulejczak
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.K.Z.); (P.R.)
| | - Anna M. Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.K.Z.); (P.R.)
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| |
Collapse
|
4
|
Li W, Gu J, Fan H, Zhang L, Guo J, Si L. Evolving cancer resistance to anti-PD-1/PD-L1 antibodies in melanoma: Comprehensive insights with future prospects. Crit Rev Oncol Hematol 2024; 201:104426. [PMID: 38908767 DOI: 10.1016/j.critrevonc.2024.104426] [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: 04/12/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024] Open
Abstract
Immunotherapy has transformed the treatment of advanced melanoma. However, up to two-thirds of patients experience disease progression after initially achieving a response to immunotherapy. Furthermore, most research has focused on cutaneous melanoma rather than acral or mucosal melanoma, although the latter predominates in Asian populations. In this review, we examine and summarize current definitions of resistance to immunotherapy and the epidemiology of resistance to PD-1 inhibition. We also review the available literature on molecular mechanisms of resistance, including how the tumor mutational landscape and tumor microenvironments of immunotherapy-resistant acral and mucosal melanomas may influence resistance. Finally, we review strategies for overcoming resistance to PD-1 inhibition and summarize completed studies and ongoing clinical trials. Our review highlights that improving the understanding of resistance mechanisms, optimizing existing therapies and further studying high-risk populations would maximize the potential of immunotherapy and result in optimized treatment outcomes for patients with melanoma.
Collapse
Affiliation(s)
- Wenyu Li
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Junjie Gu
- Department of Urological Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Hongwei Fan
- Value & Implementation, Global Medical & Scientific Affairs, Merck Sharp & Dohme (MSD) China, Shanghai, China
| | - Li Zhang
- Value & Implementation, Global Medical & Scientific Affairs, Merck Sharp & Dohme (MSD) China, Shanghai, China
| | - Jun Guo
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China; Department of Urological Oncology, Peking University Cancer Hospital and Institute, Beijing, China.
| | - Lu Si
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
5
|
Arghidash F, Javid-Naderi MJ, Gheybi F, Gholamhosseinian H, Kesharwani P, Sahebkar A. Exploring the multifaceted effects of silymarin on melanoma: Focusing on the role of lipid-based nanocarriers. J Drug Deliv Sci Technol 2024; 99:105950. [DOI: 10.1016/j.jddst.2024.105950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
Persky J, Cruz SM, Darrow MA, Judge SJ, Li Y, Bold RJ, Karnezis AN, Matsukuma KE, Qi L, Canter RJ. Characterization of natural killer and cytotoxic T-cell immune infiltrates in pancreatic ductal adenocarcinoma. J Surg Oncol 2024; 129:885-892. [PMID: 38196111 PMCID: PMC10980567 DOI: 10.1002/jso.27581] [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: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024]
Abstract
BACKGROUND AND OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with poor response to systemic therapies, including immunotherapy. Given the immunotherapeutic potential of natural killer (NK) cells, we evaluated intratumoral NK cell infiltrates along with cytotoxic T cells in PDAC to determine their association with patient outcomes. METHODS We analyzed tumors from 93 PDAC patients treated from 2012 to 2020. Predictor variables included tumor-infiltrating lymphocytes (TILs), T-cell markers (CD3, CD8, CD45RO), NK marker (NKp46), and NK inhibitory marker (major histocompatibility complex class I [MHC-I]) by immunohistochemistry. Primary outcome variables were recurrence-free survival (RFS) and overall survival (OS). RESULTS Mean TILs, CD3, and NKp46 scores were 1.3 ± 0.63, 20.6 ± 17.5, and 3.1 ± 3.9, respectively. Higher expression of CD3 and CD8 was associated with higher OS, whereas NK cell infiltration was not associated with either RFS or OS. There was a tight positive correlation between MHC-I expression and all T-cell markers, but not with NKp46. CONCLUSIONS Overall NK cell infiltrates were low in PDAC and did not predict clinical outcomes, whereas T-cell infiltrates did. Further characterization of the immune infiltrate in PDAC, including inhibitory signals and suppressive cell types, may yield better biomarkers of prognosis and immune targeting in this refractory disease.
Collapse
Affiliation(s)
- Julia Persky
- Division of Surgical Oncology, UC Davis Comprehensive Cancer Center, Sacramento, CA
| | - Sylvia M. Cruz
- Division of Surgical Oncology, UC Davis Comprehensive Cancer Center, Sacramento, CA
| | - Morgan, A. Darrow
- Department of Pathology and Laboratory Medicine, UC Davis Medical Center, Sacramento, CA
| | - Sean J. Judge
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yueju Li
- Division of Biostatistics, Department of Public Health Sciences, UC Davis
| | - Richard J. Bold
- Division of Surgical Oncology, UC Davis Comprehensive Cancer Center, Sacramento, CA
| | - Anthony N. Karnezis
- Department of Pathology and Laboratory Medicine, UC Davis Medical Center, Sacramento, CA
| | - Karen E. Matsukuma
- Department of Pathology and Laboratory Medicine, UC Davis Medical Center, Sacramento, CA
| | - Lihong Qi
- Division of Biostatistics, Department of Public Health Sciences, UC Davis
| | - Robert J. Canter
- Division of Surgical Oncology, UC Davis Comprehensive Cancer Center, Sacramento, CA
| |
Collapse
|
7
|
Filippi L, Proietti I, Petrozza V, Potenza C, Bagni O, Schillaci O. The Prognostic Role of [ 18F]FDG PET/CT in Patients with Advanced Cutaneous Squamous Cell Carcinoma Submitted to Cemiplimab Immunotherapy: A Single-Center Retrospective Study. Cancer Biother Radiopharm 2024; 39:46-54. [PMID: 37883658 DOI: 10.1089/cbr.2023.0110] [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: 10/28/2023] Open
Abstract
Background: Baseline 2-deoxy-2[18F]fluoro-d-glucose ([18F]FDG) positron emission tomography (PET)-derived parameters and 12-week metabolic response were investigated as prognostic factors in advanced cutaneous squamous cell carcinoma (cSCC) submitted to cemiplimab immunotherapy. Materials and Methods: Clinical records of 25 cSCC patients receiving cemiplimab, submitted to [18F]FDG positron emission tomography/computed tomography (PET/CT) at baseline and after ∼12 weeks, were retrospectively reviewed. The Kaplan-Meier (KM) method was applied to analyze differences in event-free survival (EFS), and Cox regression analysis was employed to identify the prognostic factors. Results: At the 12-week PET/CT evaluation, 16 patients (64%) were classified as responders (complete or partial response) and 9 (36%) as nonresponders ("unconfirmed progressive metabolic disease") according to immune PET Response Criteria in Solid Tumors (iPERCIST). By KM analysis, baseline metabolic tumor volume (MTV) and total lesion glycolysis (TLG) significantly correlated with the EFS (p < 0.05). Furthermore, the KM analysis showed that the lack of metabolic response at 12 weeks was associated with meaningfully shorter EFS (7.2 ± 1 months in nonresponders vs. 20.3 ± 2.3 months in responders). In Cox multivariate analysis, metabolic response at 12 weeks remained the only predictor of the EFS (p < 0.05). Conclusions: Baseline tumor load (i.e., MTV and TLG) and metabolic response at 12 weeks may have a prognostic impact in cSCC patients treated with cemiplimab.
Collapse
Affiliation(s)
- Luca Filippi
- Nuclear Medicine Unit, Department of Oncohaematology, Fondazione PTV Policlinico Tor Vergata University Hospital, Rome, Italy
| | - Ilaria Proietti
- Dermatology Unit "Daniele Innocenzi," "A. Fiorini" Hospital, Terracina, Italy
| | - Vincenzo Petrozza
- Department of Medico-Surgical Sciences and Biotechnologies, Pathology Unit, ICOT Hospital, University of Rome "La Sapienza," Rome, Italy
| | - Concetta Potenza
- Dermatology Unit "Daniele Innocenzi," "A. Fiorini" Hospital, Terracina, Italy
| | - Oreste Bagni
- Nuclear Medicine Unit, Santa Maria Goretti Hospital, Latina, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University Tor Vergata, Rome, Italy
| |
Collapse
|
8
|
Thomas B, Burns M, Pervanas H, Ciurescu D, Dima L. Nivolumab/Relatlimab-rmbw: A Novel Dual Combination Therapy to Treat Adult and Pediatric Patients With Unresectable or Metastatic Melanoma. Am J Ther 2023; 30:e526-e534. [PMID: 37921680 DOI: 10.1097/mjt.0000000000001680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors control effector mechanisms and work to restore downregulated T-cells in patients with melanoma. Examples of such include programmed death-1 inhibitors and lymphocyte-activating gene 3 inhibitors. The combination of nivolumab, a programmed death-1 inhibitor, and relatlimab-rmbw, a lymphocyte-activating gene 3 inhibitor, has shown antitumor activity and improved progression-free survival in patients with unresectable or metastatic melanoma. MECHANISM OF ACTION PHARMACOKINETICS/PHARMACODYNAMICS The fixed-dose combination of nivolumab and relatlimab immunotherapy is approved for adults and pediatrics 12 years of age or older with metastatic or unresectable melanoma. Volume of distribution is 6.6 L for relatlimab and nivolumab, and half-life is 27 and 26 days, respectively. Clearance at steady state is 7.6 mL/h for nivolumab and 5.5 mL/h for relatlimab. Sex, age, race, and mild hepatic/renal impairment had no clinical effect on clearance. The exposure-response relationship and pharmacodynamic response for the safety and effectiveness of nivolumab/relatlimab-rmbw have not been fully characterized. Safety concerns include severe and fatal immune-mediated adverse reactions, infusion-related reactions, and complications of allogeneic hematopoietic stem cell transplantation, and fetal toxicity. Dosing is determined by patient's age and weight. Solution is infused over a 30-minute timeframe. CLINICAL TRIALS In the RELATIVITY-047 trial, patients received nivolumab or nivolumab/relatlimab-rmbw. Results showed superiority of dual therapy over monotherapy with a progression-free survival of 10.1 months (95% CI, 6.4-15.7) compared with 4.6 months (95% CI, 3.4-5.6) and hazard ratio of 0.75 (95% CI, 0.62-0.92); P = 0.006, respectively. No safety concerns were observed compared with monotherapy with treatment-related adverse events occurring in 18.9% of patients on combination therapy compared with 9.7% on nivolumab alone. THERAPEUTIC ADVANCE The novel mechanism and improvement in progression-free survival compared with standard of care highlight the therapeutic advancement of nivolumab/relatlimab-rmbw in the treatment of unresectable and metastatic melanoma.
Collapse
Affiliation(s)
| | | | - Helen Pervanas
- Massachusetts College of Pharmacy and Health Sciences University, Manchester, NH
| | - Daniel Ciurescu
- Faculty of Medicine, Department of Medical and Surgical Specialties, Transilvania University of Brasov, Romania; and
| | - Lorena Dima
- Faculty of Medicine, Department of Fundamental Disciplines and Clinical Prevention, Transilvania University of Brasov, Brasov, Romania
| |
Collapse
|
9
|
Jiang R, Shen F, Zhang M, Mulati S, Wang J, Tao Y, Zhang W. Evaluating the Anti-Melanoma Effects and Toxicity of Cinnamaldehyde Analogues. Molecules 2023; 28:7309. [PMID: 37959729 PMCID: PMC10647553 DOI: 10.3390/molecules28217309] [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: 09/10/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Cinnamaldehyde (CA) showed potent activity against melanoma in our previous study, and the structure of unsaturated aldehydes is envisaged to play a role. Nevertheless, its limited drug availability restricts its clinical application. Therefore, a series of CA analogues were synthesized to evaluate their anti-melanoma activities across various melanoma cell lines. These compounds were also tested for their toxicity against the different normal cell lines. The compound with the most potential, CAD-14, exhibited potent activity against the A375, A875 and SK-MEL-1 cells, with IC50 values of 0.58, 0.65, and 0.82 µM, respectively. A preliminary molecular mechanism study of CAD-14 indicated that it could inhibit the p38 pathway to induce apoptosis, and suppress tumor growth by inhibiting the expression of ENO1. Furthermore, an acute toxicity study depicted that CAD-14 has better safety and tolerability than CA in vivo. These findings indicate that CAD-14 might be a lead compound for exploring effective anti-melanoma drugs.
Collapse
Affiliation(s)
- Rongsong Jiang
- School of Pharmacy, Xinjiang Medical University, Urumchi 830017, China; (R.J.); (M.Z.); (S.M.); (J.W.)
| | - Fukui Shen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China;
| | - Miaomiao Zhang
- School of Pharmacy, Xinjiang Medical University, Urumchi 830017, China; (R.J.); (M.Z.); (S.M.); (J.W.)
| | - Shulipan Mulati
- School of Pharmacy, Xinjiang Medical University, Urumchi 830017, China; (R.J.); (M.Z.); (S.M.); (J.W.)
| | - Jinfeng Wang
- School of Pharmacy, Xinjiang Medical University, Urumchi 830017, China; (R.J.); (M.Z.); (S.M.); (J.W.)
| | - Yicun Tao
- School of Pharmacy, Xinjiang Medical University, Urumchi 830017, China; (R.J.); (M.Z.); (S.M.); (J.W.)
| | - Weiyi Zhang
- School of Pharmacy, Xinjiang Medical University, Urumchi 830017, China; (R.J.); (M.Z.); (S.M.); (J.W.)
| |
Collapse
|
10
|
Redondo-Muñoz M, Rodriguez-Baena FJ, Aldaz P, Caballé-Mestres A, Moncho-Amor V, Otaegi-Ugartemendia M, Carrasco-Garcia E, Olias-Arjona A, Lasheras-Otero I, Santamaria E, Bocanegra A, Chocarro L, Grier A, Dzieciatkowska M M, Bigas C, Martin J, Urdiroz-Urricelqui U, Marzo F, Santamaria E, Kochan G, Escors D, Larrayoz IM, Heyn H, D'Alessandro A, Attolini CSO, Matheu A, Wellbrock C, Benitah SA, Sanchez-Laorden B, Arozarena I. Metabolic rewiring induced by ranolazine improves melanoma responses to targeted therapy and immunotherapy. Nat Metab 2023; 5:1544-1562. [PMID: 37563469 PMCID: PMC10513932 DOI: 10.1038/s42255-023-00861-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 07/07/2023] [Indexed: 08/12/2023]
Abstract
Resistance of melanoma to targeted therapy and immunotherapy is linked to metabolic rewiring. Here, we show that increased fatty acid oxidation (FAO) during prolonged BRAF inhibitor (BRAFi) treatment contributes to acquired therapy resistance in mice. Targeting FAO using the US Food and Drug Administration-approved and European Medicines Agency-approved anti-anginal drug ranolazine (RANO) delays tumour recurrence with acquired BRAFi resistance. Single-cell RNA-sequencing analysis reveals that RANO diminishes the abundance of the therapy-resistant NGFRhi neural crest stem cell subpopulation. Moreover, by rewiring the methionine salvage pathway, RANO enhances melanoma immunogenicity through increased antigen presentation and interferon signalling. Combination of RANO with anti-PD-L1 antibodies strongly improves survival by increasing antitumour immune responses. Altogether, we show that RANO increases the efficacy of targeted melanoma therapy through its effects on FAO and the methionine salvage pathway. Importantly, our study suggests that RANO could sensitize BRAFi-resistant tumours to immunotherapy. Since RANO has very mild side-effects, it might constitute a therapeutic option to improve the two main strategies currently used to treat metastatic melanoma.
Collapse
Grants
- P30 CA046934 NCI NIH HHS
- Ministry of Economy and Competitiveness | Instituto de Salud Carlos III (Institute of Health Carlos III)
- Departamento de Salud del Gobierno de Navarra, Spain (Grant Ref. No: GºNa 71/17)
- Marta Redondo-Muñoz is funded by a PhD studentship from the Department of Industry of the Government of Navarra, Spain. MRM acknowledges funding from the Grupo Español Multidisciplinar de Melanoma
- The University of Colorado School of Medicine Metabolomics Core is supported in part by the University of Colorado Cancer Center award from the National Cancer Institute P30CA046934
- David Escors Acknowledges funding from The Spanish Association against Cancer (AECC), PROYE16001ESCO), Biomedicine Project Grant from the Department of Health of the Government of Navarre-FEDER funds (BMED 050-2019, 51-2021) ; Strategic projects from the Department of Industry, Government of Navarre (AGATA, Ref. 0011-1411-2020-000013; LINTERNA, Ref. 0011-1411-2020-000033; DESCARTHES, 0011-1411-2019-000058).
- Research in the S.A.B. laboratory is supported partially by the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (Grant agreement No. 787041), the Government of Cataluña (SGR grant), the Government of Spain (MINECO), the La Marató/TV3 Foundation, the Foundation Lilliane Bettencourt, the Spanish Association for Cancer Research (AECC) and The Worldwide Cancer Research Foundation (WCRF)
- Work in B.S-L´s lab is funded by:PID2019-106852-RBI00 funded by MCIN/AEI/ 10.13039/501100011033, the Melanoma Research Alliance (https://doi.org/10.48050/pc.gr.91574 to B.S-L) and the FERO Foundation.
Collapse
Affiliation(s)
- Marta Redondo-Muñoz
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
| | | | - Paula Aldaz
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
| | - Adriá Caballé-Mestres
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Verónica Moncho-Amor
- Cellular Oncology Group, Biodonostia Health Research Institute, San Sebastian, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Madrid, Spain
| | | | - Estefania Carrasco-Garcia
- Cellular Oncology Group, Biodonostia Health Research Institute, San Sebastian, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Madrid, Spain
| | - Ana Olias-Arjona
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
| | - Irene Lasheras-Otero
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
| | - Eva Santamaria
- Hepatology Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Bocanegra
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Luisa Chocarro
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Abby Grier
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Monika Dzieciatkowska M
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Claudia Bigas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Josefina Martin
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Uxue Urdiroz-Urricelqui
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Florencio Marzo
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
| | - Enrique Santamaria
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Grazyna Kochan
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - David Escors
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Ignacio Marcos Larrayoz
- Biomarkers and Molecular Signaling Group, Center for Biomedical Research of La Rioja (CIBIR), Foundation Rioja Salud, Logroño, Spain
- Unidad Predepartamental de Enfermería, Universidad de La Rioja (UR), Logroño, Spain
| | - Holger Heyn
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Angelo D'Alessandro
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Camille Stephan-Otto Attolini
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Ander Matheu
- Cellular Oncology Group, Biodonostia Health Research Institute, San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Claudia Wellbrock
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
- Department of Health Sciences, Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| | | | - Imanol Arozarena
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain.
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain.
| |
Collapse
|
11
|
van Akkooi AC, Hauschild A, Long GV, Mandala M, Kicinski M, Govaerts AS, Klauck I, Ouali M, Lorigan PC, Eggermont AM. COLUMBUS-AD: phase III study of adjuvant encorafenib + binimetinib in resected stage IIB/IIC BRAF V600-mutated melanoma. Future Oncol 2023; 19:2017-2027. [PMID: 37665297 DOI: 10.2217/fon-2023-0414] [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: 09/05/2023] Open
Abstract
Stage IIB/IIC melanoma has a high risk of recurrence after surgical resection. While, for decades, surgery was the only option for high-risk stage II disease in most countries, adjuvant therapies now exist. Anti-programmed cell death protein 1 (PD-1) antibodies significantly improve recurrence-free survival versus placebo in patients with fully resected stage IIB/IIC melanoma. Combined BRAF MEK inhibitor therapy showed benefits in high-risk stage III and advanced disease; however, its role in patients with fully resected stage BRAF-mutated IIB/IIC melanoma is still unknown. Here we describe the rationale and design of the ongoing randomized, placebo-controlled COLUMBUS-AD trial, the first study of a BRAF-MEK inhibitor combination therapy (encorafenib + binimetinib) in patients with BRAF V600-mutated stage IIB/IIC melanoma.
Collapse
Affiliation(s)
- Alexander Cj van Akkooi
- Melanoma Institute Australia, the University of Sydney & Royal Prince Alfred Hospital, 40 Rocklands Road Wollstonecraft, Sydney 2065, NSW, Australia
| | - Axel Hauschild
- Department of Dermatology, University Hospital (UKSH), Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Georgina V Long
- Melanoma Institute Australia, University of Sydney, & Mater & Royal North Shore Hospitals, 40 Rocklands Road Wollstonecraft, Sydney 2065, NSW, Australia
| | - Mario Mandala
- University of Perugia, Ospedale Santa Maria della Misericordia, Piazzale Giorgio Menghini, 3, 06129, Perugia, Italy
| | - Michal Kicinski
- EORTC Headquarters, Avenue Emmanuel Mounier 83/11, 1200, Brussels, Belgium
| | | | - Isabelle Klauck
- Pierre Fabre, Medical & Patient/Consumer Division, 33 avenue Emile Zola, 92100, Boulogne-Billancourt, France
| | - Monia Ouali
- Pierre Fabre, Medical & Patient/Consumer Division, Langlade, France
| | - Paul C Lorigan
- Christie NHS Foundation Trust, Wilmslow Road Manchester M20 4BX, UK
| | - Alexander Mm Eggermont
- University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Comprehensive Cancer Center Munich, Technical University Munich & Ludwig Maximiliaan University, Marchioninistraße 15, 81377 Munich, Germany
| |
Collapse
|
12
|
Manski CF. Navigating Uncertainty in Immunotherapy Regimens. NEJM EVIDENCE 2023; 2:EVIDpp2300028. [PMID: 38320045 DOI: 10.1056/evidpp2300028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Uncertainty in Immunotherapy RegimensIn this Patient Platform, Charles Manski, Ph.D., shares his experience in navigating uncertainty from the perspective of a patient who has had to cope with a challenging clinical problem and an academic whose research aims to improve the evidence and methods used to inform patient care.
Collapse
Affiliation(s)
- Charles F Manski
- Department of Economics and Institute for Policy Research, Northwestern University, Evanston, IL
| |
Collapse
|
13
|
Augustin RC, Luke JJ. Top advances of the year: Melanoma. Cancer 2023; 129:822-828. [PMID: 36629350 PMCID: PMC11234509 DOI: 10.1002/cncr.34590] [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] [Indexed: 01/12/2023]
Abstract
This commentary highlights the key, recent advances made in the field of melanoma. Although significant gains have been made, particularly for resectable disease, ongoing challenges remain in the PD1‐refractory setting.
Collapse
Affiliation(s)
- Ryan C Augustin
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jason J Luke
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
14
|
Kaizuka T, Kurihara R, Harumiya M, Kimura T, Ushida N. Final analysis of a post-marketing surveillance study of dabrafenib and trametinib combination therapy in Japanese patients with unresectable malignant melanoma with BRAF V600 mutation. J Dermatol 2023. [PMID: 36890675 DOI: 10.1111/1346-8138.16759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 03/10/2023]
Abstract
Targeted therapy with a combination of dabrafenib and trametinib has been developed and widely used for treatment of melanoma. However, data regarding its safety and efficacy in Japanese patients with malignant melanoma are limited. A post-marketing surveillance (PMS) study was conducted to investigate the safety and efficacy of combination therapy in a Japanese clinical setting with a surveillance period of June 2016 to March 2022; 326 patients with unresectable malignant melanoma with BRAF mutation were enrolled. The interim results were published in July 2020. Herein, we report the results of the final analysis based on the data collected until the completion of the PMS study. The safety analysis population included 326 patients, the majority of whom had stage IV disease (79.14%) and Eastern Cooperative Oncology Group performance status 0 or 1 (85.28%). All patients were treated with the approved dose of dabrafenib, while 99.08% were treated with the approved dose of trametinib. Adverse events (AEs) occurred in 282 patients (86.50%) and the major AEs (incidence ≥5%) were pyrexia (47.85%), malignant melanoma (33.44%), hepatic function abnormal (9.82%), rash and blood creatine phosphokinase increased (8.59% each), malaise (6.44%), nausea (5.52%), and diarrhea and rhabdomyolysis (5.21% each). The incidences rates of adverse drug reactions of safety specifications were 45.71% for pyrexia, 15.95% for hepatic impairment, 12.58% for rhabdomyolysis, 4.60% for cardiac disorders, and 3.07% for eye disorders. In the efficacy analysis population of 318 patients, the objective response rate was 58.18% (95% confidence interval [CI] 52.54%-63.66%). The progression-free survival rates at 90, 180, and 360 days were 88.14% (95% CI 84.00%-91.26%), 69.53% (63.85%-74.50%), and 52.07% (45.71%-58.03%), respectively. Consistent with previous interim results, no new safety or efficacy concerns were observed in this final analysis of a PMS study conducted in a Japanese real-world clinical setting.
Collapse
Affiliation(s)
- Tomoaki Kaizuka
- Global Drug Development Division, Novartis Pharma K.K., Tokyo, Japan
| | - Ryohei Kurihara
- Global Drug Development Division, Novartis Pharma K.K., Tokyo, Japan
| | - Miki Harumiya
- Global Drug Development Division, Novartis Pharma K.K., Tokyo, Japan
| | - Takayuki Kimura
- Medical Affairs Department, Innovative Medicines International Japan, Novartis Pharma K.K., Tokyo, Japan
| | - Naoko Ushida
- Global Drug Development Division, Novartis Pharma K.K., Tokyo, Japan
| |
Collapse
|
15
|
Lasheras-Otero I, Feliu I, Maillo A, Moreno H, Redondo-Muñoz M, Aldaz P, Bocanegra A, Olias-Arjona A, Lecanda F, Fernandez-Irigoyen J, Santamaria E, Larrayoz IM, Gomez-Cabrero D, Wellbrock C, Vicent S, Arozarena I. The Regulators of Peroxisomal Acyl-Carnitine Shuttle CROT and CRAT Promote Metastasis in Melanoma. J Invest Dermatol 2023; 143:305-316.e5. [PMID: 36058299 DOI: 10.1016/j.jid.2022.08.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/26/2022] [Accepted: 08/03/2022] [Indexed: 01/25/2023]
Abstract
Circulating tumor cells are the key link between a primary tumor and distant metastases, but once in the bloodstream, loss of adhesion induces cell death. To identify the mechanisms relevant for melanoma circulating tumor cell survival, we performed RNA sequencing and discovered that detached melanoma cells and isolated melanoma circulating tumor cells rewire lipid metabolism by upregulating fatty acid (FA) transport and FA beta-oxidation‒related genes. In patients with melanoma, high expression of FA transporters and FA beta-oxidation enzymes significantly correlates with reduced progression-free and overall survival. Among the highest expressed regulators in melanoma circulating tumor cells were the carnitine transferases carnitine O-octanoyltransferase and carnitine acetyltransferase, which control the shuttle of peroxisome-derived medium-chain FAs toward mitochondria to fuel mitochondrial FA beta-oxidation. Knockdown of carnitine O-octanoyltransferase or carnitine acetyltransferase and short-term treatment with peroxisomal or mitochondrial FA beta-oxidation inhibitors thioridazine or ranolazine suppressed melanoma metastasis in mice. Carnitine O-octanoyltransferase and carnitine acetyltransferase depletion could be rescued by medium-chain FA supplementation, indicating that the peroxisomal supply of FAs is crucial for the survival of nonadherent melanoma cells. Our study identifies targeting the FA-based cross-talk between peroxisomes and mitochondria as a potential therapeutic opportunity to challenge melanoma progression. Moreover, the discovery of the antimetastatic activity of the Food and Drug Administration‒approved drug ranolazine carries translational potential.
Collapse
Affiliation(s)
- Irene Lasheras-Otero
- Cancer Signaling Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Iker Feliu
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Program in Solid Tumors, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Alberto Maillo
- Translational Bioinformatics Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain
| | - Haritz Moreno
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Program in Solid Tumors, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Marta Redondo-Muñoz
- Cancer Signaling Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Paula Aldaz
- Cancer Signaling Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Ana Bocanegra
- Oncoimmunology Group, Navarrabiomed, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain
| | - Ana Olias-Arjona
- Cancer Signaling Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Fernando Lecanda
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Program in Solid Tumors, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain; Center for Biomedical Research Network on Cancer (CIBERONC), Madrid, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Joaquin Fernandez-Irigoyen
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Proteomics Platform, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain
| | - Enrique Santamaria
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Clinical Neuroproteomics Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain
| | - Ignacio M Larrayoz
- Biomarkers and Molecular Signaling Group, Center for Biomedical Research of La Rioja (CIBIR), Foundation Rioja Salud, Logroño, Spain; Pre-departmental Nursing Unit, University of La Rioja (UR), Logroño, La Rioja, Spain
| | - David Gomez-Cabrero
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Translational Bioinformatics Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain; Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Claudia Wellbrock
- Cancer Signaling Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain
| | - Silvestre Vicent
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Program in Solid Tumors, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain; Center for Biomedical Research Network on Cancer (CIBERONC), Madrid, Spain
| | - Imanol Arozarena
- Cancer Signaling Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.
| |
Collapse
|
16
|
Krakowski I, Häbel H, Nielsen K, Ingvar C, Andersson TML, Girnita A, Smedby KE, Eriksson H. Association of metformin use and survival in patients with cutaneous melanoma and diabetes. Br J Dermatol 2023; 188:32-40. [PMID: 36689497 DOI: 10.1093/bjd/ljac003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/19/2022] [Accepted: 09/03/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND Metformin use has been associated with improved survival in patients with different types of cancer, but research regarding the effect of metformin on cutaneous melanoma (CM) survival is sparse and inconclusive. OBJECTIVES To investigate the association between metformin use and survival among patients with CM and diabetes. METHODS All adult patients with a primary invasive CM between 2007 and 2014 were identified in the Swedish Melanoma Registry and followed until death, or end of follow-up on 31 December 2017 in this population-based cohort study. Patients with both CM and type 2 diabetes mellitus were assessed further. Overall survival (OS) and melanoma-specific survival (MSS) were the primary endpoints. Cox proportional hazard models estimating crude and adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) were used comparing peridiagnostic use vs. nonuse of metformin. Dose response was evaluated based on defined daily doses. RESULTS Among a total of 23 507 patients, 1162 patients with CM and type 2 diabetes mellitus were included in the final cohort, with a median follow-up time of 4.1 years (interquartile range 2.4-6.1). Peridiagnostic metformin use was associated with a significantly decreased risk of death by any cause (HR 0.68, 95% CI 0.57-0.81). Cumulative pre- and postdiagnostic metformin use was also associated with improved OS: the HR for prediagnostic use was 0.90 (95% CI 0.86-0.95) for every 6 months of use and the HR for postdiagnostic use ranged from 0.98 (95% CI 0.97-0.98) for 0-6 months to 0.59 (0.49-0.70) for 24-30 months of use. No association was found for metformin use and MSS. CONCLUSIONS Metformin use was associated with improved OS in patients with CM and diabetes regardless of timing (pre-, post- or peridiagnostic use) and followed a dose-response pattern. However, further research regarding the underlying mechanisms is warranted.
Collapse
Affiliation(s)
- Isabelle Krakowski
- Department of Dermatology/Inflammation Theme
- Department of Oncology and Pathology
| | | | - Kari Nielsen
- Dermatology and Department of Dermatology, Skåne University Hospital, Lund, Sweden
| | - Christian Ingvar
- Surgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| | | | - Ada Girnita
- Department of Oncology and Pathology
- Cancer Theme, Medical Unit Head, Neck, Lung and Skin Cancer, Skin Cancer Center
| | - Karin E Smedby
- Department of Medicine Solna, Division of Clinical Epidemiology; Karolinska Institutet, Stockholm, Sweden
- Department of Hematology; Karolinska University Hospital, Stockholm, Sweden
| | - Hanna Eriksson
- Department of Oncology and Pathology
- Cancer Theme, Medical Unit Head, Neck, Lung and Skin Cancer, Skin Cancer Center
| |
Collapse
|
17
|
Wang H, Tran TT, Duong KT, Nguyen T, Le UM. Options of Therapeutics and Novel Delivery Systems of Drugs for the Treatment of Melanoma. Mol Pharm 2022; 19:4487-4505. [PMID: 36305753 DOI: 10.1021/acs.molpharmaceut.2c00775] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Melanoma is one of the most severe cancerous diseases. The cells employ multiple signaling pathways, such as ERK, HGF/c-MET, WNT, and COX-2 to cause the cell proliferation, survival, and metastasis. Treatment of melanoma, including surgery, chemotherapy, immunotherapy, radiation, and targeted therapy, is based on 4 major or 11 substages of the disease. Fourteen drugs, including dacarbazine, interferon α-2b, interleukin-12, ipilimumab, peginterferon α-2b, vemurafenib, trametinib, talimogene laherparepvec, cobimetinib, pembrolizumab, dabrafenib, binimetinib, encorafenib, and nivolumab, have been approved by the FDA for the treatment of melanoma. All of them are in conventional dosage forms of injection solutions, suspensions, oral tablets, or capsules. Major drawbacks of the treatment are side effects of the drugs and patients' incompliance to them. These are consequences of high doses and long-term treatments for the diseases. Currently more than 350 NCI-registered clinical trials are being carried out to treat advanced and/or metastatic melanoma using novel treatment methods, such as immune cell therapy, cancer vaccines, and new therapeutic targets. In addition, novel delivery systems using biomaterials of the approved drugs have been developed attempting to increase the drug delivery, targeting, stability, bioavailability, thus potentially reducing the toxicity and increasing the treatment effectiveness. Nanoparticles and liposomes have been emerging as advanced delivery systems which can improve drug stability and systemic circulation time. In this review, the most recent findings in the options for treatment and development of novel drug delivery systems for the treatment of melanoma are comprehensively discussed.
Collapse
Affiliation(s)
- Hongbin Wang
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, California 95757, United States
- Master of Pharmaceutical Sciences College of Graduate Study, California Northstate University, 9700 West Taron Drive, Elk Grove, California 95757, United States
| | - Tuan T Tran
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, California 95757, United States
| | - Katherine T Duong
- CVS Pharmacy, 18872 Beach Boulevard, Huntington Beach, California 92648, United States
| | - Trieu Nguyen
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, California 95757, United States
| | - Uyen M Le
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, California 95757, United States
| |
Collapse
|
18
|
Sun Q, Sun H, Wu N, Hu Y, Zhang F, Cong X. Patients with melanoma treated with immune checkpoint inhibitors who had non-thyroid endocrine and skin immune-related adverse events have better prognosis: A systematic review and meta-analysis. Front Oncol 2022; 12:976224. [PMID: 36185176 PMCID: PMC9515964 DOI: 10.3389/fonc.2022.976224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background Several studies have reported an association between the occurrence of immune-related adverse events (irAEs) and prognosis in patients with melanoma treated with immune checkpoint inhibitors (ICIs), but the results remain controversial. We conducted a systematic review and meta-analysis to investigate the association between irAEs and survival in patients with melanoma treated with ICIs. Methods We searched the PubMed, Web of Science, and China National Knowledge Infrastructure databases through May 5, 2022 for clinical studies evaluating the association between irAEs and in melanoma patients treated with ICIs. Combined hazard ratios (HRs) for overall survival (OS) and progression-free survival (PFS) were calculated using fixed- or random-effects models based on heterogeneity. Results A total of 60 articles were included, with 16,520 patients. In patients with melanoma treated with ICIs, the occurrence of irAEs was significantly associated with better OS (HR, 0.58; 95% confidence interval [CI], 0.51–0.66; P<0.00001) and PFS (HR, 0.61; 95%CI, 0.51–0.72; P<0.00001). Endocrine irAEs (OS, HR, 0.81; 95%CI, 0.72–0.92; P=0.001; PFS: HR, 0.84; 95%CI, 0.73–0.96, P=0.009), skin irAEs (OS, HR, 0.59; 95%CI, 0.41–0.85; P=0.004; PFS: HR, 0.43; 95%CI, 0.36–0.52; P<0.00001), vitiligo (OS, HR, 0.22; 95%CI, 0.15–0.31; P<0.00001; PFS, HR, 0.33; 95%CI, 0.25–0.44; P<0.00001), and grade 1–2 irAEs (OS, HR, 0.67; 95%CI, 0.58–0.78; P<0.00001; PFS, HR, 0.62; 95%CI, 0.51–0.76; P<0.00001) showed similar results. However, thyroid, lung, gastrointestinal, liver, and grade 3–4 irAEs were not significantly associated with OS and PFS. The occurrence of non-thyroid endocrine irAEs was significantly associated with better OS (HR, 0.22; 95%CI, 0.15–0.31; P<0.00001). In patients with melanoma treated with anti-programmed cell death protein 1 (OS, HR, 0.61; 95%CI, 0.51–0.72; P<0.00001; PFS, HR, 0.59; 95%CI, 0.47–0.74; P<0.00001), the association between irAEs and clinical benefit was clearer than in patients treated with anti-cytotoxic T-lymphocyte-associated protein 4 (OS, HR, 0.68; 95%CI, 0.52–0.89; P=0.005; PFS, HR, 0.93; 95%CI, 0.49–1.78; P=0.83). Conclusion Among patients with melanoma treated with ICIs, those who developed non-thyroid endocrine irAEs and cutaneous irAEs have better prognosis. This suggests that non-thyroid endocrine irAEs and cutaneous irAEs may be a prognostic biomarker for patients with melanoma treated with ICIs. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42022338308.
Collapse
Affiliation(s)
- Qian Sun
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hongyan Sun
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Nan Wu
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yue Hu
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Fangqing Zhang
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xianling Cong
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun, China
- *Correspondence: Xianling Cong,
| |
Collapse
|
19
|
Zhao F, Yu R, Chen S, Zhao S, Sun L, Xu Z, Zhang Y, Dai S, Zhang G, Shu Q. Global research trends on precision cancer medicine-related rashes (2008-2021): A bibliographic study. Front Immunol 2022; 13:1002034. [PMID: 36091077 PMCID: PMC9458849 DOI: 10.3389/fimmu.2022.1002034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 08/09/2022] [Indexed: 12/24/2022] Open
Abstract
BackgroundPrecision cancer medicine-related rashes are a kind of skin and mucous lesions caused by precision therapy. More and more evidences indicated that such events should not be ignored in the course of anti-tumor therapy. Since cancer treatment entered the “Precision Era”, there has been a rapid increase in this field. However, there was few bibliometric studies to provide an overall review of this field. This study aims to evaluate the literature output and trends in researches on precision cancer medicine-related rashes from a global perspective.MethodsCollected publications on precision cancer medicine-related rashes from the Web of Science Core Collection database, which were limited to articles and reviews in English. Microsoft Excel, VOS viewer and CiteSpace V were used for quantitative and visual analysis.ResultsA total of 1,229 papers were identified. From 2008 to 2021, annual publications increased year by year. The United States published the most papers in this field (44.9%) and ranking first in citation frequency (19,854 times) and H-index (69). The University of Texas system ranks first with 98 papers published. Lacouture M.E and Robert C were the principal investigators. Cancers has the largest number of articles published, with 70 articles. In recent years, there have been research hotspots related to immunotherapy, including ipilimumab, immunotherapy, tumor microenvironment, association, checkpoint inhibitor, and cutaneous adverse event.ConclusionPrecision cancer medicine-related rashes are a hot research topic in oncology. The number of relevant publications will increase dramatically. “Checkpoint inhibitors”, “skin adverse events”, “associations” and “tumor microenvironment” may become research hotspots in the future.
Collapse
Affiliation(s)
- Fangmin Zhao
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rui Yu
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuyi Chen
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuya Zhao
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lin Sun
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zeting Xu
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yao Zhang
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuying Dai
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Gaochenxi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Qijin Shu
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Qijin Shu,
| |
Collapse
|
20
|
Combination of Whole-Body Baseline CT Radiomics and Clinical Parameters to Predict Response and Survival in a Stage-IV Melanoma Cohort Undergoing Immunotherapy. Cancers (Basel) 2022; 14:cancers14122992. [PMID: 35740659 PMCID: PMC9221470 DOI: 10.3390/cancers14122992] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary The use of immunotherapeutic agents significantly improved stage-IV melanoma patients’ overall progression-free survival. To identify patients who do not benefit from immunotherapy, both clinical parameters and experimental biomarkers such as radiomics are currently being evaluated. However, no radiomic biomarker is widely accepted for routine clinical use. In a large cohort of 262 stage-IV melanoma patients given first-line immunotherapy treatment, we investigated whether radiomics—based on the segmentation of all baseline metastases in the whole body—in combination with clinical parameters offered added value compared to the usage of clinical parameters alone in a machine-learning prediction model. The primary endpoints were response at three months, and survival rates at six and twelve months. The study indicated a potential, but non-significant, added value of radiomics for six-month and twelve-month survival prediction, thus underlining the relevance of clinical parameters. Abstract Background: This study investigated whether a machine-learning-based combination of radiomics and clinical parameters was superior to the use of clinical parameters alone in predicting therapy response after three months, and overall survival after six and twelve months, in stage-IV malignant melanoma patients undergoing immunotherapy with PD-1 checkpoint inhibitors and CTLA-4 checkpoint inhibitors. Methods: A random forest model using clinical parameters (demographic variables and tumor markers = baseline model) was compared to a random forest model using clinical parameters and radiomics (extended model) via repeated 5-fold cross-validation. For this purpose, the baseline computed tomographies of 262 stage-IV malignant melanoma patients treated at a tertiary referral center were identified in the Central Malignant Melanoma Registry, and all visible metastases were three-dimensionally segmented (n = 6404). Results: The extended model was not significantly superior compared to the baseline model for survival prediction after six and twelve months (AUC (95% CI): 0.664 (0.598, 0.729) vs. 0.620 (0.545, 0.692) and AUC (95% CI): 0.600 (0.526, 0.667) vs. 0.588 (0.481, 0.629), respectively). The extended model was not significantly superior compared to the baseline model for response prediction after three months (AUC (95% CI): 0.641 (0.581, 0.700) vs. 0.656 (0.587, 0.719)). Conclusions: The study indicated a potential, but non-significant, added value of radiomics for six-month and twelve-month survival prediction of stage-IV melanoma patients undergoing immunotherapy.
Collapse
|
21
|
Jandova J, Park SL, Corenblum MJ, Madhavan L, Snell JA, Rounds L, Wondrak GT. Mefloquine induces ER stress and apoptosis in BRAFi-resistant A375-BRAF V600E /NRAS Q61K malignant melanoma cells targeting intracranial tumors in a bioluminescent murine model. Mol Carcinog 2022; 61:603-614. [PMID: 35417045 PMCID: PMC9133119 DOI: 10.1002/mc.23407] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/15/2022] [Accepted: 03/27/2022] [Indexed: 02/03/2023]
Abstract
Molecularly targeted therapeutics have revolutionized the treatment of BRAFV600E -driven malignant melanoma, but the rapid development of resistance to BRAF kinase inhibitors (BRAFi) presents a significant obstacle. The use of clinical antimalarials for the investigational treatment of malignant melanoma has shown only moderate promise, attributed mostly to inhibition of lysosomal-autophagic adaptations of cancer cells, but identification of specific antimalarials displaying single-agent antimelanoma activity has remained elusive. Here, we have screened a focused library of clinically used artemisinin-combination therapeutic (ACT) antimalarials for the apoptotic elimination of cultured malignant melanoma cell lines, also examining feasibility of overcoming BRAFi-resistance comparing isogenic melanoma cells that differ only by NRAS mutational status (BRAFi-sensitive A375-BRAFV600E /NRASQ61 vs. BRAFi-resistant A375-BRAFV600E /NRASQ61K ). Among ACT antimalarials tested, mefloquine (MQ) was the only apoptogenic agent causing melanoma cell death at low micromolar concentrations. Comparative gene expression-array analysis (A375-BRAFV600E /NRASQ61 vs. A375-BRAFV600E /NRASQ61K ) revealed that MQ is a dual inducer of endoplasmic reticulum (ER) and redox stress responses that precede MQ-induced loss of viability. ER-trackerTM DPX fluorescence imaging and electron microscopy indicated ER swelling, accompanied by rapid induction of ER stress signaling (phospho-eIF2α, XBP-1s, ATF4). Fluo-4 AM-fluorescence indicated the occurrence of cytosolic calcium overload observable within seconds of MQ exposure. In a bioluminescent murine model employing intracranial injection of A375-Luc2 (BRAFV600E /NRASQ61K ) cells, an oral MQ regimen efficiently antagonized brain tumor growth. Taken together, these data suggest that the clinical antimalarial MQ may be a valid candidate for drug repurposing aiming at chemotherapeutic elimination of malignant melanoma cells, even if metastasized to the brain and BRAFi-resistant.
Collapse
Affiliation(s)
- Jana Jandova
- Department of Pharmacology and Toxicology, RK Coit College of Pharmacy & UA Cancer Center, University of Arizona, Tucson, Arizona, USA
| | - Sophia L. Park
- Department of Pharmacology and Toxicology, RK Coit College of Pharmacy & UA Cancer Center, University of Arizona, Tucson, Arizona, USA
| | - Mandi J. Corenblum
- Department of Neurology, Evelyn F McKnight Brain Institute and BIO5 Institute, University of Arizona, Tucson, Arizona, USA
| | - Lalitha Madhavan
- Department of Neurology, Evelyn F McKnight Brain Institute and BIO5 Institute, University of Arizona, Tucson, Arizona, USA
| | - Jeremy A. Snell
- Department of Pharmacology and Toxicology, RK Coit College of Pharmacy & UA Cancer Center, University of Arizona, Tucson, Arizona, USA
| | - Liliana Rounds
- Department of Pharmacology and Toxicology, RK Coit College of Pharmacy & UA Cancer Center, University of Arizona, Tucson, Arizona, USA
| | - Georg T. Wondrak
- Department of Pharmacology and Toxicology, RK Coit College of Pharmacy & UA Cancer Center, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
22
|
Thornton J, Chhabra G, Singh CK, Guzmán-Pérez G, Shirley CA, Ahmad N. Mechanisms of Immunotherapy Resistance in Cutaneous Melanoma: Recognizing a Shapeshifter. Front Oncol 2022; 12:880876. [PMID: 35515106 PMCID: PMC9066268 DOI: 10.3389/fonc.2022.880876] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/18/2022] [Indexed: 12/21/2022] Open
Abstract
Melanoma is one of the seven most common cancers in the United States, and its incidence is still increasing. Since 2011, developments in targeted therapies and immunotherapies have been essential for significantly improving overall survival rates. Prior to the advent of targeted and immunotherapies, metastatic melanoma was considered a death sentence, with less than 5% of patients surviving more than 5 years. With the implementation of immunotherapies, approximately half of patients with metastatic melanoma now survive more than 5 years. Unfortunately, this also means that half of the patients with melanoma do not respond to current therapies and live less than 5 years after diagnosis. One major factor that contributes to lower response in this population is acquired or primary resistance to immunotherapies via tumor immune evasion. To improve the overall survival of melanoma patients new treatment strategies must be designed to minimize the risk of acquired resistance and overcome existing primary resistance. In recent years, many advances have been made in identifying and understanding the pathways that contribute to tumor immune evasion throughout the course of immunotherapy treatment. In addition, results from clinical trials focusing on treating patients with immunotherapy-resistant melanoma have reported some initial findings. In this review, we summarize important mechanisms that drive resistance to immunotherapies in patients with cutaneous melanoma. We have focused on tumor intrinsic characteristics of resistance, altered immune function, and systemic factors that contribute to immunotherapy resistance in melanoma. Exploring these pathways will hopefully yield novel strategies to prevent acquired resistance and overcome existing resistance to immunotherapy treatment in patients with cutaneous melanoma.
Collapse
Affiliation(s)
- Jessica Thornton
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Gagan Chhabra
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Chandra K Singh
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | | | - Carl A Shirley
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, WI, United States.,William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
| |
Collapse
|
23
|
Switzer B, Puzanov I, Skitzki JJ, Hamad L, Ernstoff MS. Managing Metastatic Melanoma in 2022: A Clinical Review. JCO Oncol Pract 2022; 18:335-351. [PMID: 35133862 PMCID: PMC9810138 DOI: 10.1200/op.21.00686] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cutaneous melanoma remains the most lethal of the primary cutaneous neoplasms, and although the incidence of primary melanoma continues to rise, the mortality from metastatic disease remains unchanged, in part through advances in treatment. Major developments in immunomodulatory and targeted therapies have provided robust improvements in response and survival trends that have transformed the clinical management of patients with metastatic melanoma. Additional advances in immunologic and cancer cell biology have contributed to further optimization in (1) risk stratification, (2) prognostication, (3) treatment, (4) toxicity management, and (5) surveillance approaches for patients with an advanced melanoma diagnosis. In this review, we provide a comprehensive overview of the historical and future advances regarding the translational and clinical implications of advanced melanoma and share multidisciplinary recommendations to aid clinicians in the navigation of current treatment approaches for a variety of patient cohorts.
Collapse
Affiliation(s)
- Benjamin Switzer
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Joseph J. Skitzki
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Lamya Hamad
- Department of Pharmacy, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Marc S. Ernstoff
- ImmunoOncology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD,Marc S. Ernstoff, MD, National Cancer Institute, Rockville, MD 20850; e-mail:
| |
Collapse
|
24
|
Bogéa GMR, Silva-Carvalho AÉ, Filiú-Braga LDDC, Neves FDAR, Saldanha-Araujo F. The Inflammatory Status of Soluble Microenvironment Influences the Capacity of Melanoma Cells to Control T-Cell Responses. Front Oncol 2022; 12:858425. [PMID: 35419291 PMCID: PMC8996246 DOI: 10.3389/fonc.2022.858425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/28/2022] [Indexed: 11/20/2022] Open
Abstract
The development of immunotherapeutic approaches for the treatment of melanoma requires a better understanding of immunoescape mechanisms of tumor cells and how they interact with other tumor-resident cell types. Here, we evaluated how the conditioned media of resting (rCM) and immune-activated PBMCs (iCM) influence the ability of a metastatic melanoma cell line (MeWo) to control T-cells function. MeWo cells were expanded in RPMI, rCM, or iCM and the secretome generated after cell expansion was identified as MeSec (RPMI), niSec (non-inflammatory), or iSec (inflammatory secretome), respectively. Then, the immunomodulatory potential of such secretomes was tested in PHA-activated PBMCs. iCM induced higher levels of IFN-γ and IL-10 in treated melanoma cells compared to rCM, as well as higher IDO and PD-L1 expression. The iSec was able to inhibit T-cell activation and proliferation. Interestingly, PBMCs treated with iSec presented a reduced expression of the regulators of Th1 and Th2 responses T-BET and GATA-3, as well as low expression of IFN-γ, and co-stimulatory molecules TIM-3 and LAG-3. Importantly, our findings show that melanoma may benefit from an inflammatory microenvironment to enhance its ability to control the T-cell response. Interestingly, such an immunomodulatory effect involves the inhibition of the checkpoint molecules LAG-3 and TIM-3, which are currently investigated as important therapeutic targets for melanoma treatment. Further studies are needed to better understand how checkpoint molecules are modulated by paracrine and cell contact-dependent interaction between melanoma and immune cells. Such advances are fundamental for the development of new therapeutic approaches focused on melanoma immunotherapy.
Collapse
Affiliation(s)
- Gabriela Muller Reche Bogéa
- Laboratório de Farmacologia Molecular, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
| | - Amandda Évelin Silva-Carvalho
- Laboratório de Farmacologia Molecular, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
| | - Luma Dayane de Carvalho Filiú-Braga
- Laboratório de Farmacologia Molecular, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
| | | | - Felipe Saldanha-Araujo
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
| |
Collapse
|
25
|
Devaux A, Baurain JF. Management of metastatic melanoma with new immunotherapy approaches beyond PD-1/CTLA-4 inhibitors. Curr Opin Oncol 2022; 34:123-130. [PMID: 35081051 DOI: 10.1097/cco.0000000000000821] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW If we may cure metastatic melanoma patients thanks to immune checkpoint inhibitors (ICI), it is fair to say that around 2/3 of the patients present primary or secondary resistance to ICI. Therefore, progresses are needed and numerous new treatments are tested either alone or in combination with cytolytic T-lymphocyte-associated protein 4 (CTLA-4) or (PD)-1 blockade to overcome this resistance. In this review, we focused on new immunotherapeutic approaches studied in advanced melanoma previously treated by anti-PD-1 (Programmed cell Death 1 receptor) or anti-CTLA-4 antibodies. RECENT FINDINGS The different approaches have been classified based on 'the cancer immunity cycle'. These new strategies target either the T-cell priming and activation step, T-cell trafficking and tumor infiltration, or tumor antigen recognition by T-cell and tumor killing. SUMMARY Most of these novel strategies are based on mAbs targeting T-cell inhibitory or stimulatory coreceptors. The second main focus is based on modifying the tumor micro-environment. Combination strategies seem promising in few patients and suggest that a deeper understanding of the resistance in individual patients is mandatory to go further.
Collapse
Affiliation(s)
- Alix Devaux
- Medical Oncology Department, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Bruxelles, Belgium
| | | |
Collapse
|
26
|
Prognostic Biomarkers in Uveal Melanoma: The Status Quo, Recent Advances and Future Directions. Cancers (Basel) 2021; 14:cancers14010096. [PMID: 35008260 PMCID: PMC8749988 DOI: 10.3390/cancers14010096] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/15/2021] [Accepted: 12/23/2021] [Indexed: 01/18/2023] Open
Abstract
Simple Summary Although rare, uveal melanoma (UM) is the most common cancer that develops inside adult eyes. The prognosis is poor, since 50% of patients will develop lethal metastases in the first decade, especially to the liver. Once metastases are detected, life expectancy is limited, given that the available treatments are mostly unsuccessful. Thus, there is a need to find methods that can accurately predict UM prognosis and also effective therapeutic strategies to treat this cancer. In this manuscript, we initially compile the current knowledge on epidemiological, clinical, pathological and molecular features of UM. Then, we cover the most relevant prognostic factors currently used for the evaluation and follow-up of UM patients. Afterwards, we highlight emerging molecular markers in UM published over the last three years. Finally, we discuss the problems preventing meaningful advances in the treatment and prognostication of UM patients, as well as forecast new roadblocks and paths of UM-related research. Abstract Uveal melanoma (UM) is the most common malignant intraocular tumour in the adult population. It is a rare cancer with an incidence of nearly five cases per million inhabitants per year, which develops from the uncontrolled proliferation of melanocytes in the choroid (≈90%), ciliary body (≈6%) or iris (≈4%). Patients initially present either with symptoms like blurred vision or photopsia, or without symptoms, with the tumour being detected in routine eye exams. Over the course of the disease, metastases, which are initially dormant, develop in nearly 50% of patients, preferentially in the liver. Despite decades of intensive research, the only approach proven to mildly control disease spread are early treatments directed to ablate liver metastases, such as surgical excision or chemoembolization. However, most patients have a limited life expectancy once metastases are detected, since there are limited therapeutic approaches for the metastatic disease, including immunotherapy, which unlike in cutaneous melanoma, has been mostly ineffective for UM patients. Therefore, in order to offer the best care possible to these patients, there is an urgent need to find robust models that can accurately predict the prognosis of UM, as well as therapeutic strategies that effectively block and/or limit the spread of the metastatic disease. Here, we initially summarized the current knowledge about UM by compiling the most relevant epidemiological, clinical, pathological and molecular data. Then, we revisited the most important prognostic factors currently used for the evaluation and follow-up of primary UM cases. Afterwards, we addressed emerging prognostic biomarkers in UM, by comprehensively reviewing gene signatures, immunohistochemistry-based markers and proteomic markers resulting from research studies conducted over the past three years. Finally, we discussed the current hurdles in the field and anticipated the future challenges and novel avenues of research in UM.
Collapse
|
27
|
Serum-derived exosomes promote CD8+ T cells to overexpress PD-1, affecting the prognosis of hypopharyngeal carcinoma. Cancer Cell Int 2021; 21:584. [PMID: 34717645 PMCID: PMC8557583 DOI: 10.1186/s12935-021-02294-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 10/21/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Hypopharyngeal cancer (HPC) is associated with a poor prognosis and a high recurrence rate. Immune escape is one of the reasons for the poor prognosis of malignant tumors. Programmed cell death ligand 1 (PD-L1) and programmed cell death-1 (PD-1) have been shown to play important roles in immune escape. However, the role of PD-1/PD-L1 in HPC remains unclear. In this experiment, we investigated the effect of exosomes from HPC patient serum on CD8+ T cell function and PD-1/PD-L1 expression and, thus, on prognosis. We hope to provide guidance for the identification of new targets for HPC immunotherapy. METHODS PD-1 and CD8 expression in 71 HPC tissues and 16 paracarcinoma tissues was detected by immunohistochemistry. Concurrently, the clinicopathological data of the patients were obtained to conduct correlation analysis. Exosomes were isolated from serum and then identified by Western blotting (WB), transmission electron microscopy (TEM), and nanoparticle tracking analysis (NTA). Flow cytometry was used to assess the activity of CD8+ T cells after exosome stimulation. The effects of exosomes on the ability of CD8+ T cells to kill FaDu cells were assessed by CCK-8 assay. The expression of IL-10 and TGF-β1 was measured by enzyme-linked immunosorbent assay (ELISA). PD-L1 expression in HPC tissue samples was evaluated by immunohistochemistry, and the relationship between PD-1/PD-L1 expression and prognosis was investigated with patient specimens. RESULTS PD-1 expression was significantly upregulated on CD8+ T cells in tumor tissues compared with those in normal tissues. The overall survival (OS) and disease-free survival (DFS) of PD-1-overexpressing patients were decreased. Serum exosomes from patients can elevate PD-1 expression on CD8+ T cells and suppress their killing capacity and secretory function. The rate of positive PD-L1 expression was increased in HPC tissues compared with paracancerous tissues. The DFS and OS of the PD-1(+)-PD-L1(+) group were significantly lower than those of the PD-1(-)-PD-L1(-) group. CONCLUSION Our findings indicate that serum exosomes from HPC patients can inhibit CD8+ T cell function and that the PD-1-PD-L1 pathway plays an important role in the immune escape of HPC. Exosomes combined with immunotherapy may guide the treatment of patients with advanced disease in the future.
Collapse
|
28
|
Filippi L, Proietti I, Schillaci O, Potenza C, Bagni O. A fare case of pancreatic metastasis from malignant melanoma mimicking pancreatitis on 18F-FDG PET/CT. J Egypt Natl Canc Inst 2021; 33:29. [PMID: 34632554 DOI: 10.1186/s43046-021-00087-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/04/2021] [Indexed: 11/10/2022] Open
Affiliation(s)
- Luca Filippi
- Department of Nuclear Medicine, "Santa Maria Goretti" Hospital, Via Antonio Canova, 04100, Latina, Italy.
| | - Ilaria Proietti
- Dermatology Unit "Daniele Innocenzi", "A. Fiorini" Hospital, Via Firenze, 1, 04019, Terracina, LT, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University Tor Vergata, Viale Oxford 81, 00133, Rome, Italy.,IRCCS Neuromed, 86077, Pozzilli, Italy
| | - Concetta Potenza
- Dermatology Unit "Daniele Innocenzi", "A. Fiorini" Hospital, Via Firenze, 1, 04019, Terracina, LT, Italy
| | - Oreste Bagni
- Department of Nuclear Medicine, "Santa Maria Goretti" Hospital, Via Antonio Canova, 04100, Latina, Italy
| |
Collapse
|
29
|
Becker AL, Carpenter EL, Slominski AT, Indra AK. The Role of the Vitamin D Receptor in the Pathogenesis, Prognosis, and Treatment of Cutaneous Melanoma. Front Oncol 2021; 11:743667. [PMID: 34692525 PMCID: PMC8526885 DOI: 10.3389/fonc.2021.743667] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/15/2021] [Indexed: 01/08/2023] Open
Abstract
Melanoma is the malignant transformation of melanocytes and represents the most lethal form of skin cancer. While early-stage melanoma localized to the skin can be cured with surgical excision, metastatic melanoma often requires a multi-pronged approach and even then can exhibit treatment resistance. Understanding the molecular mechanisms involved in the pathogenesis of melanoma could lead to novel diagnostic, prognostic, and therapeutic strategies to ultimately decrease morbidity and mortality. One emerging candidate that may have value as both a prognostic marker and in a therapeutic context is the vitamin D receptor (VDR). VDR is a nuclear steroid hormone receptor activated by 1,25 dihydroxy-vitamin D3 [calcitriol, 1,25(OH)2D3]. While 1,25 dihydroxy-vitamin D3 is typically thought of in relation to calcium metabolism, it also plays an important role in cell proliferation, differentiation, programmed-cell death as well as photoprotection. This review discusses the role of VDR in the crosstalk between keratinocytes and melanocytes during melanomagenesis and summarizes the clinical data regarding VDR polymorphisms, VDR as a prognostic marker, and potential uses of vitamin D and its analogs as an adjuvant treatment for melanoma.
Collapse
Affiliation(s)
- Alyssa L. Becker
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, OR, United States
- John A. Burns School of Medicine at the University of Hawai’i at Mānoa, Honolulu, HI, United States
| | - Evan L. Carpenter
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, OR, United States
| | - Andrzej T. Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, United States
- Cancer Chemoprevention Program, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Arup K. Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, OR, United States
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR, United States
- Department of Biochemistry and Biophysics, Oregon State University (OSU), Corvallis, OR, United States
- Linus Pauling Science Center, Oregon State University (OSU), Corvallis, OR, United States
- Department of Dermatology, Oregon Health & Science University (OHSU), Portland, OR, United States
| |
Collapse
|